1
|
Mustan F, Genchev N, Vinarova L, Bevernage J, Tistaert C, Ivanova A, Tcholakova S, Vinarov Z. Understanding drug solubilization in intestinal mixed micelles through molecular dynamics simulations. J Colloid Interface Sci 2025; 684:225-234. [PMID: 39827542 DOI: 10.1016/j.jcis.2025.01.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
HYPOTHESIS Solubilization is a fundamental process that underpins various technologies in the pharmaceutical and chemical industry. However, knowledge of the location, orientation and interactions of solubilized molecules in the micelles is still limited. We expect all-atom molecular dynamics simulations to improve the molecular-level understanding of solubilization and to enable its in silico prediction. METHODS The solubilization of six drugs in intestinal mixed micelles composed of taurocholate and dioleoyl phosphatidylcholine was simulated by molecular dynamics in explicit water and measured experimentally by liquid chromatography. The location and orientation of the solubilized drugs were visualized by cumulative radial distribution functions and interactions were characterized by radial distribution function ratios and hydrogen bonding. FINDINGS A new simulation-derived parameter was defined, which accounts for drug-micelle and drug-water interactions and correlates (R2 = 0.83) with the experimentally measured solubilization. Lipophilicity was found to govern the location of all drugs in the micelle (hydrophobic core, palisade layer or on the surface), while hydrogen bonding was crucial for orientation and solubilization of two of the molecules. The study demonstrates that explicit, hydrogen bond-forming water molecules are vital for accurate prediction of solubilization and provides a comprehensive framework for quantitative studies of drug location and orientation within the micelles.
Collapse
Affiliation(s)
- Fatmegyul Mustan
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, University of Sofia, Bulgaria.
| | - Nikola Genchev
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, University of Sofia, Bulgaria
| | - Liliya Vinarova
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, University of Sofia, Bulgaria
| | | | | | - Anela Ivanova
- Department of Physical Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia, Bulgaria
| | - Slavka Tcholakova
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, University of Sofia, Bulgaria
| | - Zahari Vinarov
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, University of Sofia, Bulgaria
| |
Collapse
|
2
|
Hernández-Martín M, Garcimartín A, Bocanegra A, Macho-González A, García-Fernández RA, de Pascual-Teresa S, Redondo-Castillejo R, Bastida S, Sánchez-Muniz FJ, Benedí J, López-Oliva ME. Silicon-Enriched Meat Ameliorates Diabetic Dyslipidemia by Improving Cholesterol, Bile Acid Metabolism and Ileal Barrier Integrity in Rats with Late-Stage Type 2 Diabetes. Int J Mol Sci 2024; 25:11405. [PMID: 39518958 PMCID: PMC11547133 DOI: 10.3390/ijms252111405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Silicon as a functional ingredient of restructured meat (RM) shows antidiabetic and hypocholesterolemic effects in a type 2 diabetes mellitus (T2DM) rat model. The present paper investigated the mechanisms involved in this cholesterol-lowering effect by studying the impact of silicon-RM consumption on bile acid (BA) and cholesterol metabolism. In addition, the main effects of cecal BA and short-chain fatty acids derived from the microbiota on intestinal barrier integrity were also tested. Rats were fed an RM high-saturated-fat, high-cholesterol diet (HSFHCD) combined with a low dose of streptozotocin plus nicotinamide injection (LD group) and for an 8 wk. period. Silicon-RM was included in the HSFHCD as a functional food (LD-Si group). An early-stage T2DM group fed a high-saturated-fat diet (ED group) was used as a reference. Silicon decreased the BA pool with a higher hydrophilic BA profile and a lower ability to digest fat and decreased the damaging effects, increasing the occludin levels and the integrity of the intestinal barrier. The ileal BA uptake and hepatic BA synthesis through CYP7A1 were reduced by FXR/FGF15 signaling activation. The silicon up-regulated the hepatic and ileal FXR and LXRα/β, improving transintestinal cholesterol (TICE), biliary BA and cholesterol effluxes. The inclusion of silicon in meat products could be used as a new therapeutic nutritional tool in the treatment of diabetic dyslipidemia.
Collapse
Affiliation(s)
- Marina Hernández-Martín
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain;
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
| | - Alba Garcimartín
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Aránzazu Bocanegra
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Adrián Macho-González
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
- Nutrition and Food Science Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rosa A. García-Fernández
- Animal Medicine and Surgery Department, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Sonia de Pascual-Teresa
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN-CSIC), 28040 Madrid, Spain;
| | - Rocío Redondo-Castillejo
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Sara Bastida
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
- Nutrition and Food Science Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Francisco J. Sánchez-Muniz
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
- Nutrition and Food Science Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Juana Benedí
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Mª Elvira López-Oliva
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain;
- AFUSAN Research Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain; (A.G.); (A.B.); (A.M.-G.); (R.R.-C.); (S.B.); (F.J.S.-M.); (J.B.)
| |
Collapse
|
3
|
Zhang Y, Chen Z, Xiao Y, Wu T, Yang H, Liu Y, Zhou R, Xiong Y, Xiong Y, Yang X, Zhou J, Zhou H, Zhang W, Shu Y, Li X, Guo F, Yin J, Liao S, Li Q, Zhu P. Effects of Compound Probiotics on Pharmacokinetics of Cytochrome 450 Probe Drugs in Rats. Drug Metab Dispos 2024; 52:1297-1312. [PMID: 39214665 DOI: 10.1124/dmd.124.001837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Compound probiotics have been widely used and commonly coadministered with other drugs for treating various chronic illnesses, yet their effects on drug pharmacokinetics remain underexplored. This study elucidated the impact of VSL#3 on the metabolism of probe drugs for cytochrome P450 enzymes (P450s), specifically omeprazole, tolbutamide, midazolam, metoprolol, phenacetin, and chlorzoxazone. Male Wistar rats were administered drinking water containing VSL#3 or not for 14 days and then intragastrically administered a P450 probe cocktail; this was done to investigate the host P450's metabolic phenotype. Stool, liver/jejunum, and serum samples were collected for 16S ribosomal RNA sequencing, RNA sequencing, and bile acid profiling. The results indicated significant differences in both α and β diversity of intestinal microbial composition between the probiotic and vehicle groups in rats. In the probiotic group, the bioavailability of omeprazole increased by 269.9%, whereas those of tolbutamide and chlorpropamide decreased by 28.1% and 27.4%, respectively. The liver and jejunum exhibited 1417 and 4004 differentially expressed genes, respectively, between the two groups. In the probiotic group, most of P450 genes were upregulated in the liver but downregulated in the jejunum. The expression of genes encoding metabolic enzymes and drug transporters also changed. The serum-conjugated bile acids in the probiotic group were significantly reduced. Shorter duodenal villi and longer ileal villi were found in the probiotic group. In summary, VSL#3 administration altered the gut microbiota, host drug-processing gene expression, and intestinal structure in rats, which could be reasons for pharmacokinetic changes. SIGNIFICANCE STATEMENT: This study focused on the effects of the probiotic VSL#3 on the pharmacokinetic profile of cytochrome P450 probe drugs and the expression of host drug metabolism genes. Compared with previous studies, the present study provides a comprehensive explanation for the host drug metabolism profile modified by probiotics, combined here with the bile acid profile and histopathological analysis.
Collapse
Affiliation(s)
- Yanjuan Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Zhi Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yayi Xiao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Tianyuan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Haijun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Rong Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yalan Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yanling Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Xuechun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Jian Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yan Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Xiong Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Fugang Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Jianhui Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Shang Liao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Peng Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| |
Collapse
|
4
|
Steigert S, Brouwers J, Verbeke K, Vanuytsel T, Augustijns P. Characterization of luminal contents from the fasted human proximal colon. Eur J Pharm Sci 2024; 200:106821. [PMID: 38823599 DOI: 10.1016/j.ejps.2024.106821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/03/2024]
Abstract
To treat colonic diseases more effectively, improved therapies are urgently needed. In this respect, delivering drugs locally to the colon is a key strategy to achieve higher local drug concentrations while minimizing systemic side effects. Understanding the luminal environment is crucial to efficiently develop such targeted therapies and to predict drug disposition in the colon. In this clinical study, we collected colonic contents from an undisturbed fasted proximal colon via colonoscopy and characterized their composition with regard to drug disposition. Colonic pH, osmolality, protein content, bile salts, lipids, phospholipids and short-chain fatty acids were investigated in 10 healthy volunteers (8 male and 2 female, age 19-25). The unique environment of the proximal colon was reflected in the composition of the sampled luminal fluids and the effect of the microbiota could be observed on the pH (median 6.55), the composition of bile salts (majority deconjugated and secondary), and the abundance of short-chain fatty acids. At the same time, an increase in phospholipid concentration, osmolality and total protein content compared to reported ileal values was seen, likely resulting from desiccation. Lipids could only be found in low quantities and mainly in the form of cholesterol and free fatty acids, showing almost complete digestion and absorption by the time luminal contents reach the colon. All characteristics also displayed the considerable intersubject variability found in different regions of the gastrointestinal tract. This study contributes to an improved understanding of the luminal conditions in the proximal colon and facilitates the development of new predictive tools to study colonic drug absorption.
Collapse
Affiliation(s)
- Sebastian Steigert
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - box 921, 3000 Leuven, Belgium
| | - Joachim Brouwers
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - box 921, 3000 Leuven, Belgium
| | - Kristin Verbeke
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Gastroenterology and Hepatology, University Hospitals Leuven campus Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | - Patrick Augustijns
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - box 921, 3000 Leuven, Belgium.
| |
Collapse
|
5
|
Verdegaal AA, Goodman AL. Integrating the gut microbiome and pharmacology. Sci Transl Med 2024; 16:eadg8357. [PMID: 38295186 PMCID: PMC12121898 DOI: 10.1126/scitranslmed.adg8357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
The gut microbiome harbors trillions of organisms that contribute to human health and disease. These bacteria can also affect the properties of medical drugs used to treat these diseases, and drugs, in turn, can reshape the microbiome. Research addressing interdependent microbiome-host-drug interactions thus has broad impact. In this Review, we discuss these interactions from the perspective of drug bioavailability, absorption, metabolism, excretion, toxicity, and drug-mediated microbiome modulation. We survey approaches that aim to uncover the mechanisms underlying these effects and opportunities to translate this knowledge into new strategies to improve the development, administration, and monitoring of medical drugs.
Collapse
Affiliation(s)
- Andrew A. Verdegaal
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Andrew L. Goodman
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| |
Collapse
|
6
|
de Waal T, Brouwers J, Rayyan M, Stillhart C, Vinarova L, Vinarov Z, Augustijns P. Characterization of neonatal and infant enterostomy fluids - Part II: Drug solubility. Int J Pharm 2023:123141. [PMID: 37321462 DOI: 10.1016/j.ijpharm.2023.123141] [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: 04/25/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
Previous research revealed marked differences in the composition of intestinal fluids between infants and adults. To explore the impact on the solubilization of orally administered drugs, the present study assessed the solubility of five poorly water-soluble, lipophilic drugs in intestinal fluid pools from 19 infant enterostomy patients (infant HIF). For some but not all drugs, the average solubilizing capacity of infant HIF was similar to that of HIF obtained from adults (adult HIF) in fed conditions. Commonly used fed state simulated intestinal fluids (FeSSIF(-V2)) predicted fairly well drug solubility in the aqueous fraction of infant HIF, but did not account for the substantial solubilization by the lipid phase of infant HIF. Despite similarities in the average solubilities of some drugs in infant HIF and adult HIF or SIF, the underlying solubilization mechanisms likely differ, considering important compositional differences (e.g., low bile salt levels). Finally, the huge variability in composition of infant HIF pools resulted in a highly variable solubilizing capacity, potentially causing variations in drug bioavailability. The current study warrants future research focusing on (i) understanding the mechanisms underlying drug solubilization in infant HIF and (ii) evaluating the sensitivity of oral drug products to interpatient variations in drug solubilization.
Collapse
Affiliation(s)
- Tom de Waal
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Joachim Brouwers
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Maissa Rayyan
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | | | - Liliya Vinarova
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University, Sofia, Bulgaria
| | - Zahari Vinarov
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University, Sofia, Bulgaria
| | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
7
|
de Waal T, Brouwers J, Berben P, Flanagan T, Tack J, Vandenberghe W, Vanuytsel T, Augustijns P. Characterization of Aspirated Duodenal Fluids from Parkinson's Disease Patients. Pharmaceutics 2023; 15:pharmaceutics15041243. [PMID: 37111729 PMCID: PMC10145225 DOI: 10.3390/pharmaceutics15041243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/16/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Parkinson's disease, one of the most common neurodegenerative diseases, may not only affect the motor system, but also the physiology of the gastrointestinal tract. Delayed gastric emptying, impaired motility and altered intestinal bacteria are well-established consequences of the disease, which can have a pronounced effect on the absorption of orally administered drugs. In contrast, no studies have been performed into the composition of intestinal fluids. It is not unlikely that Parkinson's disease also affects the composition of intestinal fluids, a critical factor in the in vitro and in silico simulation of drug dissolution, solubilization and absorption. In the current study, duodenal fluids were aspirated from Parkinson's disease (PD) patients and age-matched healthy controls (healthy controls, HC) consecutively in fasted and fed conditions. The fluids were then characterized for pH, buffer capacity, osmolality, total protein, phospholipids, bile salts, cholesterol and lipids. In a fasted state, the intestinal fluid composition was highly similar in PD patients and healthy controls. In general, the same was true for fed-state fluids, apart from a slightly slower and less pronounced initial change in factors directly affected by the meal (i.e., buffer capacity, osmolality, total protein and lipids) in PD patients. The absence of a fast initial increase for these factors immediately after meal intake, as was observed in healthy controls, might result from slower gastric emptying in PD patients. Irrespective of the prandial state, a higher relative amount of secondary bile salts was observed in PD patients, potentially indicating altered intestinal bacterial metabolism. Overall, the data from this study indicate that only minor disease-specific adjustments in small intestinal fluid composition should be considered when simulating intestinal drug absorption in PD patients.
Collapse
Affiliation(s)
- Tom de Waal
- Drug Delivery and Disposition, KU Leuven, 3000 Leuven, Belgium
| | | | - Philippe Berben
- Pharmaceutical Sciences, UCB Pharma SA, 1420 Braine-l'Alleud, Belgium
| | - Talia Flanagan
- Pharmaceutical Sciences, UCB Pharma SA, 1420 Braine-l'Alleud, Belgium
| | - Jan Tack
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, 3000 Leuven, Belgium
| | - Wim Vandenberghe
- Department of Neurology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Tim Vanuytsel
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, 3000 Leuven, Belgium
| | | |
Collapse
|
8
|
de Waal T, Brouwers J, Mols R, Hoffman I, Rayyan M, Augustijns P. Characterization of neonatal and infant enterostomy fluids. Int J Pharm 2023; 639:122943. [PMID: 37059240 DOI: 10.1016/j.ijpharm.2023.122943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/16/2023]
Abstract
The composition of gastrointestinal (GI) fluids is crucial for the dissolution, solubilization, and absorption of orally administered drugs. Disease- or age-related changes in GI fluid composition could significantly affect the pharmacokinetics of oral drugs. However, limited studies have been conducted on the characteristics of GI fluids in neonates and infants due to practical and ethical challenges. The current study collected enterostomy fluids from 21 neonate and infant patients over an extended period of time and from different regions of the small intestine and colon. The fluids were characterized for pH, buffer capacity, osmolality, total protein, bile salts, phospholipids, cholesterol, and lipid digestion products. The study found a large variability in the fluid characteristics among the different patients, in line with the highly heterogeneous study population. Compared to adult intestinal fluids, the enterostomy fluids from neonates and infants had low bile salt concentrations, with an increasing trend as a function of age; no secondary bile salts were detected. In contrast, total protein and lipid concentrations were relatively high, even in the distal small intestine. These findings suggest marked differences in intestinal fluid composition between neonates and infants versus adults, which may affect the absorption of certain drugs.
Collapse
Affiliation(s)
- Tom de Waal
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | | | - Raf Mols
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Ilse Hoffman
- Paediatric Gastroenterology. Hepatology and Nutrition, University Hospitals Leuven, Leuven, Belgium
| | - Maissa Rayyan
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | | |
Collapse
|
9
|
Yang H, Zhang Y, Zhou R, Wu T, Zhu P, Liu Y, Zhou J, Xiong Y, Xiong Y, Zhou H, Zhang W, Shu Y, Li X, Li Q. Antibiotics-Induced Depletion of Rat Microbiota Induces Changes in the Expression of Host Drug-Processing Genes and Pharmacokinetic Behaviors of CYPs Probe Drugs. Drug Metab Dispos 2023; 51:509-520. [PMID: 36623881 DOI: 10.1124/dmd.122.001173] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
The metabolism of exogenous substances is affected by the gut microbiota, and the relationship between them has become a hot topic. However, the mechanisms by which the microbiota regulates drug metabolism have not been clearly defined. This study characterizes the expression profiles of host drug-processing genes (DPGs) in antibiotics-treated rats by using an unbias quantitative RNA-sequencing method and investigates the effects of antibiotics-induced depletion of rat microbiota on the pharmacokinetic behaviors of cytochrome P450s (CYPs) probe drugs, and bile acids metabolism by ultra-performance liquid chromatography-tandem mass spectrometry. Our results show that antibiotics treatments altered the mRNA expressions of 112 DPGs in the liver and jejunum of rats. The mRNA levels of CYP2A1, CYP2C11, CYP2C13, CYP2D, CYP2E1, and CYP3A of CYP family members were significantly downregulated in antibiotics-treated rats. Furthermore, antibiotics treatments also resulted in a significant decrease in the protein expressions and enzyme activities of CYP3A1 and CYP2E1 in rat liver. Pharmacokinetic results showed that, except for tolbutamide, antibiotics treatments significantly altered the pharmacokinetic behaviors of phenacetin, omeprazole, metoprolol, chlorzoxazone, and midazolam. In conclusion, the presence of stable, complex, and diverse gut microbiota plays a significant role in regulating the expression of host DPGs, which could contribute to some individual differences in pharmacokinetics. SIGNIFICANCE STATEMENT: This study investigated how the depletion of rat microbiota by antibiotics treatments influences the expression profiles of host DPGs and the pharmacokinetic behaviors of CYPs probe drugs. Combined with previous studies in germ-free mice, this study will improve the understanding of the role of gut microbiota in drug metabolism and contribute to the understanding of individual differences in the pharmacokinetics of some drugs.
Collapse
Affiliation(s)
- Haijun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yanjuan Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Rong Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Tianyuan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Peng Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Jian Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yalan Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yanling Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yan Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Xiong Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., X.L., Q.L.); National Clinical Research Center for Geriatric Disorders, Changsha, China (H.Y., Y.Z., R.Z., T.W., P.Z., Y.L., J.Z., Yalan X., Yanling X., H.Z., W.Z., Q.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| |
Collapse
|
10
|
Amin N, Schwarzkopf S, Tröscher-Mußotter J, Camarinha-Silva A, Dänicke S, Huber K, Frahm J, Seifert J. Host metabolome and faecal microbiome shows potential interactions impacted by age and weaning times in calves. Anim Microbiome 2023; 5:12. [PMID: 36788596 PMCID: PMC9926800 DOI: 10.1186/s42523-023-00233-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Calves undergo nutritional, metabolic, and behavioural changes from birth to the entire weaning period. An appropriate selection of weaning age is essential to reduce the negative effects caused by weaning-related dietary transitions. This study monitored the faecal microbiome and plasma metabolome of 59 female Holstein calves during different developmental stages and weaning times (early vs. late) and identified the potential associations of the measured parameters over an experimental period of 140 days. RESULTS A progressive development of the microbiome and metabolome was observed with significant differences according to the weaning groups (weaned at 7 or 17 weeks of age). Faecal samples of young calves were dominated by bifidobacterial and lactobacilli species, while their respective plasma samples showed high concentrations of amino acids (AAs) and biogenic amines (BAs). However, as the calves matured, the abundances of potential fiber-degrading bacteria and the plasma concentrations of sphingomyelins (SMs), few BAs and acylcarnitines (ACs) were increased. Early-weaning at 7 weeks significantly restructured the microbiome towards potential fiber-degrading bacteria and decreased plasma concentrations of most of the AAs and SMs, few BAs and ACs compared to the late-weaning event. Strong associations between faecal microbes, plasma metabolites and calf growth parameters were observed during days 42-98, where the abundances of Bacteroides, Parabacteroides, and Blautia were positively correlated with the plasma concentrations of AAs, BAs and SMs as well as the live weight gain or average daily gain in calves. CONCLUSION The present study reported that weaning at 17 weeks of age was beneficial due to higher growth rate of late-weaned calves during days 42-98 and a quick adaptability of microbiota to weaning-related dietary changes during day 112, suggesting an age-dependent maturation of the gastrointestinal tract. However, the respective plasma samples of late-weaned calves contained several metabolites with differential concentrations to the early-weaned group, suggesting a less abrupt but more-persistent effect of dietary changes on host metabolome compared to the microbiome.
Collapse
Affiliation(s)
- Nida Amin
- grid.9464.f0000 0001 2290 1502HoLMiR - Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Stuttgart, Germany ,grid.9464.f0000 0001 2290 1502Institute of Animal Science, University of Hohenheim, Emil-Wolff-Str. 6-10, 70593 Stuttgart, Germany
| | - Sarah Schwarzkopf
- grid.9464.f0000 0001 2290 1502HoLMiR - Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Stuttgart, Germany ,grid.9464.f0000 0001 2290 1502Institute of Animal Science, University of Hohenheim, Emil-Wolff-Str. 6-10, 70593 Stuttgart, Germany
| | - Johanna Tröscher-Mußotter
- grid.9464.f0000 0001 2290 1502HoLMiR - Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Stuttgart, Germany ,grid.9464.f0000 0001 2290 1502Institute of Animal Science, University of Hohenheim, Emil-Wolff-Str. 6-10, 70593 Stuttgart, Germany
| | - Amélia Camarinha-Silva
- grid.9464.f0000 0001 2290 1502HoLMiR - Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Stuttgart, Germany ,grid.9464.f0000 0001 2290 1502Institute of Animal Science, University of Hohenheim, Emil-Wolff-Str. 6-10, 70593 Stuttgart, Germany
| | - Sven Dänicke
- grid.417834.dInstitute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Brunswick, Germany
| | - Korinna Huber
- grid.9464.f0000 0001 2290 1502HoLMiR - Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Stuttgart, Germany ,grid.9464.f0000 0001 2290 1502Institute of Animal Science, University of Hohenheim, Emil-Wolff-Str. 6-10, 70593 Stuttgart, Germany
| | - Jana Frahm
- grid.417834.dInstitute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Brunswick, Germany
| | - Jana Seifert
- HoLMiR - Hohenheim Center for Livestock Microbiome Research, University of Hohenheim, Stuttgart, Germany. .,Institute of Animal Science, University of Hohenheim, Emil-Wolff-Str. 6-10, 70593, Stuttgart, Germany.
| |
Collapse
|
11
|
Đanić M, Pavlović N, Lazarević S, Stanimirov B, Vukmirović S, Al-Salami H, Mooranian A, Mikov M. Bioaccumulation and biotransformation of simvastatin in probiotic bacteria: A step towards better understanding of drug-bile acids-microbiome interactions. Front Pharmacol 2023; 14:1111115. [PMID: 36843926 PMCID: PMC9946981 DOI: 10.3389/fphar.2023.1111115] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction: Although pharmacogenetics and pharmacogenomics have been at the forefront of research aimed at finding novel personalized therapies, the focus of research has recently extended to the potential of intestinal microbiota to affect drug efficacy. Complex interplay of gut microbiota with bile acids may have significant repercussions on drug pharmacokinetics. However, far too little attention has been paid to the potential implication of gut microbiota and bile acids in simvastatin response which is characterized by large interindividual variations. The Aim: In order to gain more insight into the underlying mechanism and its contribution in assessing the clinical outcome, the aim of our study was to examine simvastatin bioaccumulation and biotransformation in probiotic bacteria and the effect of bile acids on simvastatin bioaccumulation in in vitro conditions. Materials and methods: Samples with simvastatin, probiotic bacteria and three different bile acids were incubated at anaerobic conditions at 37°C for 24 h. Extracellular and intracellular medium samples were collected and prepared for the LC-MS analysis at predetermined time points (0 min, 15 min, 1 h, 2 h, 4 h, 6 h, 24 h). The concentrations of simvastatin were analyzed by LC-MS/MS. Potential biotransformation pathways were analyzed using a bioinformatics approach in correlation with experimental assay. Results: During the incubation, simvastatin was transported into bacteria cells leading to a drug bioaccumulation over the time, which was augmented upon addition of bile acids after 24 h. A decrease of total drug level during the incubation indicates that the drug is partly biotransformed by bacterial enzymes. According to the results of bioinformatics analysis, the lactone ring is the most susceptible to metabolic changes and the most likely reactions include ester hydrolysis followed by hydroxylation. Conclusion: Results of our study reveal that bioaccumulation and biotransformation of simvastatin by intestinal bacteria might be the underlying mechanisms of altered simvastatin bioavailability and therapeutic effect. Since this study is based only on selected bacterial strains in vitro, further more in-depth research is needed in order to elicit completely the contribution of complex drug-microbiota-bile acids interactions to overall clinical response of simvastatin which could ultimately lead to novel approaches for the personalized lipid-lowering therapy.
Collapse
Affiliation(s)
- Maja Đanić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Nebojša Pavlović
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Slavica Lazarević
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia,*Correspondence: Slavica Lazarević,
| | - Bojan Stanimirov
- Department of Biochemistry, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Saša Vukmirović
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia,Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, WA, Australia
| | - Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia,Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, WA, Australia
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
12
|
Zhou J, Ouyang J, Gao Z, Qin H, Jun W, Shi T. MagMD: database summarizing the Metabolic action of gut Microbiota to Drugs. Comput Struct Biotechnol J 2022; 20:6427-6430. [DOI: 10.1016/j.csbj.2022.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
|
13
|
Gawałko M, Agbaedeng TA, Saljic A, Müller DN, Wilck N, Schnabel R, Penders J, Rienstra M, van Gelder I, Jespersen T, Schotten U, Crijns HJGM, Kalman JM, Sanders P, Nattel S, Dobrev D, Linz D. Gut microbiota, dysbiosis and atrial fibrillation. Arrhythmogenic mechanisms and potential clinical implications. Cardiovasc Res 2022; 118:2415-2427. [PMID: 34550344 PMCID: PMC9400433 DOI: 10.1093/cvr/cvab292] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/08/2021] [Accepted: 07/25/2021] [Indexed: 02/06/2023] Open
Abstract
Recent preclinical and observational cohort studies have implicated imbalances in gut microbiota composition as a contributor to atrial fibrillation (AF). The gut microbiota is a complex and dynamic ecosystem containing trillions of microorganisms, which produces bioactive metabolites influencing host health and disease development. In addition to host-specific determinants, lifestyle-related factors such as diet and drugs are important determinants of the gut microbiota composition. In this review, we discuss the evidence suggesting a potential bidirectional association between AF and gut microbiota, identifying gut microbiota-derived metabolites as possible regulators of the AF substrate. We summarize the effect of gut microbiota on the development and progression of AF risk factors, including heart failure, hypertension, obesity, and coronary artery disease. We also discuss the potential anti-arrhythmic effects of pharmacological and diet-induced modifications of gut microbiota composition, which may modulate and prevent the progression to AF. Finally, we highlight important gaps in knowledge and areas requiring future investigation. Although data supporting a direct relationship between gut microbiota and AF are very limited at the present time, emerging preclinical and clinical research dealing with mechanistic interactions between gut microbiota and AF is important as it may lead to new insights into AF pathophysiology and the discovery of novel therapeutic targets for AF.
Collapse
Affiliation(s)
- Monika Gawałko
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
- Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen, Duisburg, Germany
- Department of Cardiology, Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A Agbaedeng
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dominik N Müller
- Experimental and Clinical Research Center, Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Max Delbrück Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center, Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Max Delbrück Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Renate Schnabel
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - John Penders
- Department of Medical Microbiology, Care and Public Health Research Institute (CAPHRI), School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Michiel Rienstra
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Isabelle van Gelder
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrich Schotten
- Department of Physiology, University Maastricht, Maastricht, The Netherlands
| | - Harry J G M Crijns
- Department of Cardiology, Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Jonathan M Kalman
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Stanley Nattel
- Department of Pharmacology, Medicine and Research Centre, Montréal Heart Institute, University de Montréal, McGill University, Montréal, QC, Canada
- IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen, Duisburg, Germany
- Department of Pharmacology, Medicine and Research Centre, Montréal Heart Institute, University de Montréal, McGill University, Montréal, QC, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Dominik Linz
- Department of Cardiology, Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
- Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
14
|
Ferreira WT, Hong HA, Adams JRG, Hess M, Kotowicz NK, Tan S, Ferrari E, Brisson A, Zentek J, Soloviev M, Cutting SM. Environmentally Acquired Bacillus and Their Role in C. difficile Colonization Resistance. Biomedicines 2022; 10:930. [PMID: 35625667 PMCID: PMC9138776 DOI: 10.3390/biomedicines10050930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 01/27/2023] Open
Abstract
Clostridioides difficile is an environmentally acquired, anaerobic, spore-forming bacterium which ordinarily causes disease following antibiotic-mediated dysbiosis of the intestinal microbiota. Although much is understood regarding the life cycle of C. difficile, the fate of C. difficile spores upon ingestion remains unclear, and the underlying factors that predispose an individual to colonization and subsequent development of C. difficile infection (CDI) are not fully understood. Here, we show that Bacillus, a ubiquitous and environmentally acquired, spore-forming bacterium is associated with colonization resistance to C. difficile. Using animal models, we first provide evidence that animals housed under conditions that mimic reduced environmental exposure have an increased susceptibility to CDI, correlating with a loss in Bacillus. Lipopeptide micelles (~10 nm) produced by some Bacilli isolated from the gastro-intestinal (GI)-tract and shown to have potent inhibitory activity to C. difficile have recently been reported. We show here that these micelles, that we refer to as heterogenous lipopeptide lytic micelles (HELMs), act synergistically with components present in the small intestine to augment inhibitory activity against C. difficile. Finally, we show that provision of HELM-producing Bacillus to microbiota-depleted animals suppresses C. difficile colonization thereby demonstrating the significant role played by Bacillus in colonization resistance. In the wider context, our study further demonstrates the importance of environmental microbes on susceptibility to pathogen colonization.
Collapse
Affiliation(s)
- William T. Ferreira
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (W.T.F.); (H.A.H.); (J.R.G.A.); (M.H.)
| | - Huynh A. Hong
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (W.T.F.); (H.A.H.); (J.R.G.A.); (M.H.)
| | - James R. G. Adams
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (W.T.F.); (H.A.H.); (J.R.G.A.); (M.H.)
| | - Mateusz Hess
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (W.T.F.); (H.A.H.); (J.R.G.A.); (M.H.)
| | - Natalia K. Kotowicz
- SporeGen Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK;
| | - Sisareuth Tan
- Laboratoire d’Imagerie Moléculaire et Nano-Bio-Technologie, UMR-CBMN CNRS-Université de Bordeaux-IPB, 33607 Pessac, France; (S.T.); (A.B.)
| | - Enrico Ferrari
- School of Life Sciences, University of Lincoln, Lincoln LN6 7TS, UK;
| | - Alain Brisson
- Laboratoire d’Imagerie Moléculaire et Nano-Bio-Technologie, UMR-CBMN CNRS-Université de Bordeaux-IPB, 33607 Pessac, France; (S.T.); (A.B.)
| | - Jurgen Zentek
- Institute for Animal Health, Freie University of Berlin, Berlin 14195, Germany;
| | - Mikhail Soloviev
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (W.T.F.); (H.A.H.); (J.R.G.A.); (M.H.)
| | - Simon M. Cutting
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK; (W.T.F.); (H.A.H.); (J.R.G.A.); (M.H.)
- SporeGen Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK;
| |
Collapse
|
15
|
Way GW, Jackson KG, Muscu SR, Zhou H. Key Signaling in Alcohol-Associated Liver Disease: The Role of Bile Acids. Cells 2022; 11:1374. [PMID: 35456053 PMCID: PMC9031669 DOI: 10.3390/cells11081374] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 02/01/2023] Open
Abstract
Alcohol-associated liver disease (ALD) is a spectrum of diseases, the onset and progression of which are due to chronic alcohol use. ALD ranges, by increasing severity, from hepatic steatosis to alcoholic hepatitis (AH) and alcohol-associated cirrhosis (AC), and in some cases, can lead to the development of hepatocellular carcinoma (HCC). ALD continues to be a significant health burden and is now the main cause of liver transplantations in the United States. ALD leads to biological, microbial, physical, metabolic, and inflammatory changes in patients that vary depending on disease severity. ALD deaths have been increasing in recent years and are projected to continue to increase. Current treatment centers focus on abstinence and symptom management, with little in the way of resolving disease progression. Due to the metabolic disruption and gut dysbiosis in ALD, bile acid (BA) signaling and metabolism are also notably affected and play a prominent role in disease progression in ALD, as well as other liver disease states, such as non-alcoholic fatty liver disease (NAFLD). In this review, we summarize the recent advances in the understanding of the mechanisms by which alcohol consumption induces hepatic injury and the role of BA-mediated signaling in the pathogenesis of ALD.
Collapse
Affiliation(s)
- Grayson W. Way
- Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Kaitlyn G. Jackson
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; (K.G.J.); (S.R.M.)
| | - Shreya R. Muscu
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; (K.G.J.); (S.R.M.)
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; (K.G.J.); (S.R.M.)
- Central Virginia Veterans Healthcare System, Richmond, VA 23249, USA
| |
Collapse
|
16
|
Wilson CG, Aarons L, Augustijns P, Brouwers J, Darwich AS, De Waal T, Garbacz G, Hansmann S, Hoc D, Ivanova A, Koziolek M, Reppas C, Schick P, Vertzoni M, García-Horsman JA. Integration of advanced methods and models to study drug absorption and related processes: An UNGAP perspective. Eur J Pharm Sci 2021; 172:106100. [PMID: 34936937 DOI: 10.1016/j.ejps.2021.106100] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023]
Abstract
This collection of contributions from the European Network on Understanding Gastrointestinal Absorption-related Processes (UNGAP) community assembly aims to provide information on some of the current and newer methods employed to study the behaviour of medicines. It is the product of interactions in the immediate pre-Covid period when UNGAP members were able to meet and set up workshops and to discuss progress across the disciplines. UNGAP activities are divided into work packages that cover special treatment populations, absorption processes in different regions of the gut, the development of advanced formulations and the integration of food and pharmaceutical scientists in the food-drug interface. This involves both new and established technical approaches in which we have attempted to define best practice and highlight areas where further research is needed. Over the last months we have been able to reflect on some of the key innovative approaches which we were tasked with mapping, including theoretical, in silico, in vitro, in vivo and ex vivo, preclinical and clinical approaches. This is the product of some of us in a snapshot of where UNGAP has travelled and what aspects of innovative technologies are important. It is not a comprehensive review of all methods used in research to study drug dissolution and absorption, but provides an ample panorama of current and advanced methods generally and potentially useful in this area. This collection starts from a consideration of advances in a priori approaches: an understanding of the molecular properties of the compound to predict biological characteristics relevant to absorption. The next four sections discuss a major activity in the UNGAP initiative, the pursuit of more representative conditions to study lumenal dissolution of drug formulations developed independently by academic teams. They are important because they illustrate examples of in vitro simulation systems that have begun to provide a useful understanding of formulation behaviour in the upper GI tract for industry. The Leuven team highlights the importance of the physiology of the digestive tract, as they describe the relevance of gastric and intestinal fluids on the behaviour of drugs along the tract. This provides the introduction to microdosing as an early tool to study drug disposition. Microdosing in oncology is starting to use gamma-emitting tracers, which provides a link through SPECT to the next section on nuclear medicine. The last two papers link the modelling approaches used by the pharmaceutical industry, in silico to Pop-PK linking to Darwich and Aarons, who provide discussion on pharmacometric modelling, completing the loop of molecule to man.
Collapse
Affiliation(s)
- Clive G Wilson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, Glasgow, U.K.
| | | | | | | | | | | | | | | | | | | | - Mirko Koziolek
- NCE Formulation Sciences, Abbvie Deutschland GmbH & Co. KG, Germany
| | | | - Philipp Schick
- Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, Germany
| | | | | |
Collapse
|
17
|
Shu X, Li M, Cao Y, Li C, Zhou W, Ji G, Zhang L. Berberine Alleviates Non-alcoholic Steatohepatitis Through Modulating Gut Microbiota Mediated Intestinal FXR Activation. Front Pharmacol 2021; 12:750826. [PMID: 34603061 PMCID: PMC8484326 DOI: 10.3389/fphar.2021.750826] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
Berberine is a natural plant alkaloid isolated from a diverse range of genera, it obtains anti-inflammatory, anti-obesity, and hepatoprotective properties, and is a promising agent for non-alcoholic steatohepatitis (NASH). Farnesoid X receptor (FXR) is a bile acid receptor and a drug target for NASH, however, the underlying mechanisms of berberine on regulating FXR are still unknown. In the present study, we feed mice with a 12-week high-fat diet with interval dextran sulfate sodium (0.5% in drinking water) diet to induce NASH, and treat the mice with berberine (100 mg/kg per day) via oral gavage for additional 4 weeks. We demonstrate that administration of berberine alleviates steatosis and infiltration of inflammatory cells in the liver of NASH mice. We apply 16S ribosomal DNA sequencing to screen the structure of gut microbiota, and ultra-performance liquid chromatography-tandem mass spectrometry analysis to determine the bile acid profiles. The results show that berberine modulates gut dysbiosis, and specifically increases the relative abundance of Clostridiales, Lactobacillaceae, and Bacteroidale. Berberine modulated microbiomes are associated with bile acid de-conjugation and transformation, which are consistent with the altered bile acid species (e.g., deoxycholic acid, ursodeoxycholic acid) upon berberine treatment. BA species that respond to berberine treatment are known FXR agonists, thus we performed quantitative Real Time-PCR and western blot to examine the FXR pathway, and find that berberine up-regulates intestinal FXR and fibroblast growth factor 15 (FGF15) expression, and the secretion of FGF15 further inhibits lipogenesis and nuclear factor-κB activation in the liver. Whereas the beneficial effects of berberine are blunted in FXR knockout mice. Our results reveal that berberine alleviates NASH by modulating the interplay of gut microbiota and bile acid metabolism, as well as the subsequent intestinal FXR activation.
Collapse
Affiliation(s)
- Xiangbing Shu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Geratology, Baoshan Branch of Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Cao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunlin Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
18
|
McCoubrey LE, Gaisford S, Orlu M, Basit AW. Predicting drug-microbiome interactions with machine learning. Biotechnol Adv 2021; 54:107797. [PMID: 34260950 DOI: 10.1016/j.biotechadv.2021.107797] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Pivotal work in recent years has cast light on the importance of the human microbiome in maintenance of health and physiological response to drugs. It is now clear that gastrointestinal microbiota have the metabolic power to promote, inactivate, or even toxify the efficacy of a drug to a level of clinically relevant significance. At the same time, it appears that drug intake has the propensity to alter gut microbiome composition, potentially affecting health and response to other drugs. Since the precise composition of an individual's microbiome is unique, one's drug-microbiome relationship is similarly unique. Thus, in the age of evermore personalised medicine, the ability to predict individuals' drug-microbiome interactions is highly sought. Machine learning (ML) offers a powerful toolkit capable of characterising and predicting drug-microbiota interactions at the individual patient level. ML techniques have the potential to learn the mechanisms operating drug-microbiome activities and measure patients' risk of such occurrences. This review will outline current knowledge at the drug-microbiota interface, and present ML as a technique for examining and forecasting personalised drug-microbiome interactions. When harnessed effectively, ML could alter how the pharmaceutical industry and healthcare professionals consider the drug-microbiome axis in patient care.
Collapse
Affiliation(s)
| | | | - Mine Orlu
- University College London, London, United Kingdom
| | | |
Collapse
|
19
|
Charpentier J, Briand F, Lelouvier B, Servant F, Azalbert V, Puel A, Christensen JE, Waget A, Branchereau M, Garret C, Lluch J, Heymes C, Brousseau E, Burcelin R, Guzylack L, Sulpice T, Grasset E. Liraglutide targets the gut microbiota and the intestinal immune system to regulate insulin secretion. Acta Diabetol 2021; 58:881-897. [PMID: 33723651 DOI: 10.1007/s00592-020-01657-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/09/2020] [Indexed: 01/05/2023]
Abstract
AIMS Liraglutide controls type 2 diabetes (T2D) and inflammation. Gut microbiota regulates the immune system and causes at least in part type 2 diabetes. We here evaluated whether liraglutide regulates T2D through both gut microbiota and immunity in dysmetabolic mice. METHODS Diet-induced dysmetabolic mice were treated for 14 days with intraperitoneal injection of liraglutide (100 µg/kg) or with vehicle or Exendin 4 (10 µg/kg) as controls. Various metabolic parameters, the intestinal immune cells were characterized and the 16SrDNA gene sequenced from the gut. The causal role of gut microbiota was shown using large spectrum antibiotics and by colonization of germ-free mice with the gut microbiota from treated mice. RESULTS Besides, the expected metabolic impacts liraglutide treatment induced a specific gut microbiota specific signature when compared to vehicle or Ex4-treated mice. However, liraglutide only increased glucose-induced insulin secretion, reduced the frequency of Th1 lymphocytes, and increased that of TReg in the intestine. These effects were abolished by a concomitant antibiotic treatment. Colonization of germ-free mice with gut microbiota from liraglutide-treated diabetic mice improved glucose-induced insulin secretion and regulated the intestinal immune system differently from what observed in germ-free mice colonized with microbiota from non-treated diabetic mice. CONCLUSIONS Altogether, our result demonstrated first the influence of liraglutide on gut microbiota and the intestinal immune system which could at least in part control glucose-induced insulin secretion.
Collapse
Affiliation(s)
- Julie Charpentier
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Francois Briand
- PHYSIOGENEX SAS Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Benjamin Lelouvier
- Vaiomer, Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Florence Servant
- Vaiomer, Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Vincent Azalbert
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Anthony Puel
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Jeffrey E Christensen
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Aurélie Waget
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Maxime Branchereau
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Céline Garret
- Vaiomer, Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Jérome Lluch
- Vaiomer, Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Christophe Heymes
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Emmanuel Brousseau
- PHYSIOGENEX SAS Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France.
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France.
| | - Laurence Guzylack
- Neuro-Gastroenterology and Nutrition Team, Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Thierry Sulpice
- PHYSIOGENEX SAS Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Estelle Grasset
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| |
Collapse
|
20
|
Zhang X, Han Y, Huang W, Jin M, Gao Z. The influence of the gut microbiota on the bioavailability of oral drugs. Acta Pharm Sin B 2021; 11:1789-1812. [PMID: 34386321 PMCID: PMC8343123 DOI: 10.1016/j.apsb.2020.09.013] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/27/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Due to its safety, convenience, low cost and good compliance, oral administration attracts lots of attention. However, the efficacy of many oral drugs is limited to their unsatisfactory bioavailability in the gastrointestinal tract. One of the critical and most overlooked factors is the symbiotic gut microbiota that can modulate the bioavailability of oral drugs by participating in the biotransformation of oral drugs, influencing the drug transport process and altering some gastrointestinal properties. In this review, we summarized the existing research investigating the possible relationship between the gut microbiota and the bioavailability of oral drugs, which may provide great ideas and useful instructions for the design of novel drug delivery systems or the achievement of personalized medicine.
Collapse
Key Words
- 5-ASA, 5-aminosalicylic acid
- AA, ascorbic acid
- ABC, ATP-binding cassette
- ACS, amphipathic chitosan derivative
- AMI, amiodarone
- AQP4, aquaporin 4
- AR, azoreductase
- ASP, amisulpride
- BBR, berberine
- BCRP, breast cancer resistance protein
- BCS, biopharmaceutics classification system
- BDDCS, the biopharmaceutics drug disposition classification system
- BDEPT, the bacteria-directed enzyme prodrug therapy
- BSH, bile salt hydrolase
- Bioavailability
- CA, cholic acid
- CDCA, chenodeoxycholic acid
- CPP, cell-penetrating peptide
- CS, chitosan
- Colon-specific drug delivery system
- DCA, deoxycholic acid
- DRPs, digoxin reduction products
- EcN, Escherichia coli Nissle 1917
- FA, folate
- FAO, Food and Agriculture Organization of the United Nations
- GCDC, glycochenodeoxycholate
- GL, glycyrrhizic acid
- Gut microbiota
- HFD, high fat diet
- HTC, hematocrit
- IBD, inflammatory bowel disease
- LCA, lithocholic acid
- LPS, lipopolysaccharide
- MATEs, multidrug and toxin extrusion proteins
- MDR1, multidrug resistance gene 1
- MDR1a, multidrug resistance protein-1a
- MKC, monoketocholic acid
- MPA, mycophenolic acid
- MRP2, multidrug resistance-associated protein 2
- NEC, necrotizing enterocolitis
- NMEs, new molecular entities
- NRs, nitroreductases
- NSAIDs, non-steroidal anti-inflammatory drugs
- NaDC, sodium deoxycholate
- NaGC, sodium glycholate
- OATs, organic anion transporters
- OCTNs, organic zwitterion/cation
- OCTs, organic cation transporters
- Oral drugs
- P-gp, P-glycoprotein
- PD, Parkinson's disease
- PPIs, proton pump inhibitors
- PT, pectin
- PWSDs, poorly water-soluble drugs
- Probiotics
- RA, rheumatoid arthritis
- RBC, red blood cell
- SCFAs, short-chain fatty acids
- SGLT-1, sodium-coupled glucose transporter 1
- SLC, solute carrier
- SLN, solid lipid nanoparticle
- SP, sulfapyridine
- SSZ, sulfasalazine
- SVCT-1/2, the sodium-dependent vitamin C transporter-1/2
- T1D, type 1 diabetes
- T1DM, type 1 diabetes mellitus
- T2D, type 2 diabetes
- TCA, taurocholate
- TCDC, taurochenodeoxycholate
- TDCA, taurodeoxycholate
- TLCA, taurolithocholate
- TME, the tumor microenvironment
- UDC, ursodeoxycholic acid
- WHO, World Health Organization
- an OTC drug, an over-the-counter drug
- cgr operon, cardiac glycoside reductase operon
- dhBBR, dihydroberberine
- pKa, dissociation constant
- the GI tract, the gastrointestinal tract
Collapse
Affiliation(s)
- Xintong Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
21
|
Li DD, Yu P, Xiao W, Wang ZZ, Zhao LG. Berberine: A Promising Natural Isoquinoline Alkaloid for the Development of Hypolipidemic Drugs. Curr Top Med Chem 2021; 20:2634-2647. [PMID: 32901585 DOI: 10.2174/1568026620666200908165913] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 05/28/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022]
Abstract
Berberine, as a representative isoquinoline alkaloid, exhibits significant hypolipidemic activity in both animal models and clinical trials. Recently, a large number of studies on the lipid-lowering mechanism of berberine and studies for improving its hypolipidemic activity have been reported, but for the most part, they have been either incomplete or not comprehensive. In addition, there have been a few specific reviews on the lipid-reducing effect of berberine. In this paper, the physicochemical properties, the lipid-lowering mechanism, and studies of the modification of berberine all are discussed to promote the development of berberine as a lipid-lowering agent. Subsequently, this paper provides some insights into the deficiencies of berberine in the study of lipid-lowering drug, and based on the situation, some proposals are put forward.
Collapse
Affiliation(s)
- Dong-Dong Li
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China,College of Chemical Engineering, Nanjing Forestry University, 159 Long Pan Road, Nanjing 210037, China
| | - Pan Yu
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China,College of Chemical Engineering, Nanjing Forestry University, 159 Long Pan Road, Nanjing 210037, China
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical Co., Ltd., 58 Haichang South Road, Lianyungang 222001, China
| | - Zhen-Zhong Wang
- Jiangsu Kanion Pharmaceutical Co., Ltd., 58 Haichang South Road, Lianyungang 222001, China
| | - Lin-Guo Zhao
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China,College of Chemical Engineering, Nanjing Forestry University, 159 Long Pan Road, Nanjing 210037, China
| |
Collapse
|
22
|
Spichak S, Bastiaanssen TFS, Berding K, Vlckova K, Clarke G, Dinan TG, Cryan JF. Mining microbes for mental health: Determining the role of microbial metabolic pathways in human brain health and disease. Neurosci Biobehav Rev 2021; 125:698-761. [PMID: 33675857 DOI: 10.1016/j.neubiorev.2021.02.044] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
There is increasing knowledge regarding the role of the microbiome in modulating the brain and behaviour. Indeed, the actions of microbial metabolites are key for appropriate gut-brain communication in humans. Among these metabolites, short-chain fatty acids, tryptophan, and bile acid metabolites/pathways show strong preclinical evidence for involvement in various aspects of brain function and behaviour. With the identification of neuroactive gut-brain modules, new predictive tools can be applied to existing datasets. We identified 278 studies relating to the human microbiota-gut-brain axis which included sequencing data. This spanned across psychiatric and neurological disorders with a small number also focused on normal behavioural development. With a consistent bioinformatics pipeline, thirty-five of these datasets were reanalysed from publicly available raw sequencing files and the remainder summarised and collated. Among the reanalysed studies, we uncovered evidence of disease-related alterations in microbial metabolic pathways in Alzheimer's Disease, schizophrenia, anxiety and depression. Amongst studies that could not be reanalysed, many sequencing and technical limitations hindered the discovery of specific biomarkers of microbes or metabolites conserved across studies. Future studies are warranted to confirm our findings. We also propose guidelines for future human microbiome analysis to increase reproducibility and consistency within the field.
Collapse
Affiliation(s)
- Simon Spichak
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Thomaz F S Bastiaanssen
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Kirsten Berding
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Klara Vlckova
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland.
| |
Collapse
|
23
|
Gvoic M, Vukmirovic S, Al-Salami H, Mooranian A, Mikov M, Stankov K. Bile acids as novel enhancers of CNS targeting antitumor drugs: a comprehensive review. Pharm Dev Technol 2021; 26:617-633. [PMID: 33882793 DOI: 10.1080/10837450.2021.1916032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite a relatively low prevalence of primary brain tumors, they continuously attract scientific interest because of the complexity of their treatment due to their location behind the blood-brain barrier. The main challenge in treatment of brain tumors is not the efficacy of the drugs, per se, but the low efficiency of drug delivery to malignant cells. At the core of the problem is the complex structure of the blood-brain barrier. Nowadays, there is evidence supporting the claim that bile acids have the ability to cross the blood-brain barrier. That ability can be exploited by taking a part in novel drug carrier designs. Bile acids represent a drug carrier system as a part of a mixed micelle composition, bilosomes and conjugates with various drugs. This review discusses the current knowledge related to bile acid molecules as drug penetration modifying agents, with the focus on central nervous system antitumor drug delivery.
Collapse
Affiliation(s)
- Marija Gvoic
- Department of Pharmacology and Toxicology and Clinical Pharmacology, Medical faculty of Novi Sad, University of Novi sad, Novi Sad, Serbia
| | - Sasa Vukmirovic
- Department of Pharmacology and Toxicology and Clinical Pharmacology, Medical faculty of Novi Sad, University of Novi sad, Novi Sad, Serbia
| | - Hani Al-Salami
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Armin Mooranian
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Momir Mikov
- Department of Pharmacology and Toxicology and Clinical Pharmacology, Medical faculty of Novi Sad, University of Novi sad, Novi Sad, Serbia
| | - Karmen Stankov
- Department of Biochemistry, Medical faculty of Novi Sad, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
24
|
Vinarov Z, Abrahamsson B, Artursson P, Batchelor H, Berben P, Bernkop-Schnürch A, Butler J, Ceulemans J, Davies N, Dupont D, Flaten GE, Fotaki N, Griffin BT, Jannin V, Keemink J, Kesisoglou F, Koziolek M, Kuentz M, Mackie A, Meléndez-Martínez AJ, McAllister M, Müllertz A, O'Driscoll CM, Parrott N, Paszkowska J, Pavek P, Porter CJH, Reppas C, Stillhart C, Sugano K, Toader E, Valentová K, Vertzoni M, De Wildt SN, Wilson CG, Augustijns P. Current challenges and future perspectives in oral absorption research: An opinion of the UNGAP network. Adv Drug Deliv Rev 2021; 171:289-331. [PMID: 33610694 DOI: 10.1016/j.addr.2021.02.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Although oral drug delivery is the preferred administration route and has been used for centuries, modern drug discovery and development pipelines challenge conventional formulation approaches and highlight the insufficient mechanistic understanding of processes critical to oral drug absorption. This review presents the opinion of UNGAP scientists on four key themes across the oral absorption landscape: (1) specific patient populations, (2) regional differences in the gastrointestinal tract, (3) advanced formulations and (4) food-drug interactions. The differences of oral absorption in pediatric and geriatric populations, the specific issues in colonic absorption, the formulation approaches for poorly water-soluble (small molecules) and poorly permeable (peptides, RNA etc.) drugs, as well as the vast realm of food effects, are some of the topics discussed in detail. The identified controversies and gaps in the current understanding of gastrointestinal absorption-related processes are used to create a roadmap for the future of oral drug absorption research.
Collapse
Affiliation(s)
- Zahari Vinarov
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Department of Chemical and Pharmaceutical Engineering, Sofia University, Sofia, Bulgaria
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Philippe Berben
- Pharmaceutical Development, UCB Pharma SA, Braine- l'Alleud, Belgium
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - James Butler
- GlaxoSmithKline Research and Development, Ware, United Kingdom
| | | | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Gøril Eide Flaten
- Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | | | | | | | | | - Martin Kuentz
- Institute for Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, Basel, Switzerland
| | - Alan Mackie
- School of Food Science & Nutrition, University of Leeds, Leeds, United Kingdom
| | | | | | - Anette Müllertz
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Petr Pavek
- Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
| | | | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Kiyohiko Sugano
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Elena Toader
- Faculty of Medicine, University of Medicine and Pharmacy of Iasi, Romania
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saskia N De Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clive G Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
25
|
Impact of gastrointestinal tract variability on oral drug absorption and pharmacokinetics: An UNGAP review. Eur J Pharm Sci 2021; 162:105812. [PMID: 33753215 DOI: 10.1016/j.ejps.2021.105812] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/19/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
The absorption of oral drugs is frequently plagued by significant variability with potentially serious therapeutic consequences. The source of variability can be traced back to interindividual variability in physiology, differences in special populations (age- and disease-dependent), drug and formulation properties, or food-drug interactions. Clinical evidence for the impact of some of these factors on drug pharmacokinetic variability is mounting: e.g. gastric pH and emptying time, small intestinal fluid properties, differences in pediatrics and the elderly, and surgical changes in gastrointestinal anatomy. However, the link of colonic factors variability (transit time, fluid composition, microbiome), sex differences (male vs. female) and gut-related diseases (chronic constipation, anorexia and cachexia) to drug absorption variability has not been firmly established yet. At the same time, a way to decrease oral drug pharmacokinetic variability is provided by the pharmaceutical industry: clinical evidence suggests that formulation approaches employed during drug development can decrease the variability in oral exposure. This review outlines the main drivers of oral drug exposure variability and potential approaches to overcome them, while highlighting existing knowledge gaps and guiding future studies in this area.
Collapse
|
26
|
Loisios-Konstantinidis I, Dressman J. Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling to Support Waivers of In Vivo Clinical Studies: Current Status, Challenges, and Opportunities. Mol Pharm 2020; 18:1-17. [PMID: 33320002 DOI: 10.1021/acs.molpharmaceut.0c00903] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling has been extensively applied to quantitatively translate in vitro data, predict the in vivo performance, and ultimately support waivers of in vivo clinical studies. In the area of biopharmaceutics and within the context of model-informed drug discovery and development (MID3), there is a rapidly growing interest in applying verified and validated mechanistic PBPK models to waive in vivo clinical studies. However, the regulatory acceptance of PBPK analyses for biopharmaceutics and oral drug absorption applications, which is also referred to variously as "PBPK absorption modeling" [Zhang et al. CPT: Pharmacometrics Syst. Pharmacol. 2017, 6, 492], "physiologically based absorption modeling", or "physiologically based biopharmaceutics modeling" (PBBM), remains rather low [Kesisoglou et al. J. Pharm. Sci. 2016, 105, 2723] [Heimbach et al. AAPS J. 2019, 21, 29]. Despite considerable progress in the understanding of gastrointestinal (GI) physiology, in vitro biopharmaceutic and in silico tools, PBPK models for oral absorption often suffer from an incomplete understanding of the physiology, overparameterization, and insufficient model validation and/or platform verification, all of which can represent limitations to their translatability and predictive performance. The complex interactions of drug substances and (bioenabling) formulations with the highly dynamic and heterogeneous environment of the GI tract in different age, ethnic, and genetic groups as well as disease states have not been yet fully elucidated, and they deserve further research. Along with advancements in the understanding of GI physiology and refinement of current or development of fully mechanistic in silico tools, we strongly believe that harmonization, interdisciplinary interaction, and enhancement of the translational link between in vitro, in silico, and in vivo will determine the future of PBBM. This Perspective provides an overview of the current status of PBBM, reflects on challenges and knowledge gaps, and discusses future opportunities around PBPK/PD models for oral absorption of small and large molecules to waive in vivo clinical studies.
Collapse
Affiliation(s)
| | - Jennifer Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main 60438, Germany.,Fraunhofer Institute of Translational Pharmacology and Medicine (ITMP), Carl-von-Noorden Platz 9, Frankfurt am Main 60438, Germany
| |
Collapse
|
27
|
Pawar G, Papadatou-Soulou E, Mason J, Muhammed R, Watson A, Cotter C, Abdallah M, Harrad S, Mackie C, Arien T, Inghelbrecht S, Batchelor H. Characterisation of fasted state gastric and intestinal fluids collected from children. Eur J Pharm Biopharm 2020; 158:156-165. [PMID: 33259897 DOI: 10.1016/j.ejpb.2020.11.010] [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] [Received: 09/17/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 10/22/2022]
Abstract
Fundamental knowledge about the composition of intestinal fluids in paediatric populations is currently unavailable. This study aimed to characterise gastric and intestinal fluid from paediatric populations. Gastric and intestinal fluid samples were obtained during routine clinical endoscopy from paediatric patients at a large teaching hospital. These fluids were characterised to measure the pH; buffer capacity; osmolality; bile acid concentration and composition. A total of 55 children were recruited to the study aged from 11 months to 15 years of age where 53 gastric fluid samples and 40 intestinal fluid samples were obtained. pH values recorded ranged from pH 0.57 to 11.05 (median: 2.50) in gastric fluids and from 0.89 to 8.97 (median: 3.27) in intestinal fluids. The buffer capacity did not change significantly between gastric and intestinal fluids with median values of 12 mM/L/ΔpH for both fluids. Gastric fluid osmolality values ranged from 1 to 615 mOsm/kg, while intestinal fluid values ranged from 35 to 631 mOsm/kg. Gastric fluid bile acid concentrations ranged from 0.002 to 2.3 mM with a median value of 0.017 mM whilst intestinal fluid bile acid concentrations ranged from 0.0008 to 3.3 mM with a median value of 0.178 mM. Glycocholate; taurocholic acid; glycochenodeoxycholate and taurochenodeoxycholate were the most commonly identified bile acids within paediatric intestinal fluids. All compositional components were associated with large inter-individual variability. Further work is required to develop simulated paediatric media and to explore the impact of these media on drug solubility and dissolution.
Collapse
Affiliation(s)
- Gopal Pawar
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Eleni Papadatou-Soulou
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Julie Mason
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Rafeeq Muhammed
- Birmingham Children's Hospital, Steelhouse Lane, Birmingham B4 6NH, United Kingdom
| | - Alison Watson
- Birmingham Children's Hospital, Steelhouse Lane, Birmingham B4 6NH, United Kingdom
| | - Catherine Cotter
- Birmingham Children's Hospital, Steelhouse Lane, Birmingham B4 6NH, United Kingdom
| | - Mohamed Abdallah
- School of Geography, Earth, and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Analytical Chemistry, Faculty of Pharmacy, Assiut University, 71526 Assiut, Egypt
| | - Stuart Harrad
- School of Geography, Earth, and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Claire Mackie
- Janssen Research and Development, Turnhoutseweg 34, 2340 Beerse, Belgium
| | - Tina Arien
- Janssen Research and Development, Turnhoutseweg 34, 2340 Beerse, Belgium
| | | | - Hannah Batchelor
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, United Kingdom.
| |
Collapse
|
28
|
Development and evaluation of a biorelevant medium simulating porcine gastrointestinal fluids. Eur J Pharm Biopharm 2020; 154:116-126. [DOI: 10.1016/j.ejpb.2020.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/08/2020] [Accepted: 06/14/2020] [Indexed: 12/23/2022]
|
29
|
Coombes Z, Yadav V, E. McCoubrey L, Freire C, W. Basit A, Conlan RS, Gonzalez D. Progestogens Are Metabolized by the Gut Microbiota: Implications for Colonic Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12080760. [PMID: 32806503 PMCID: PMC7464400 DOI: 10.3390/pharmaceutics12080760] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023] Open
Abstract
Following oral administration, the bioavailability of progestogens is very low and highly variable, in part due to metabolism by cytochrome P450 enzymes found in the mucosa of the small intestine. Conversely, the mucosa in the colon contains much lower levels of cytochrome P450 enzymes, thus, colonic delivery of progestogens may be beneficial. Microbiota in the colon are known to metabolize a great number of drugs, therefore, it is important to understand the stability of these hormones in the presence of colonic flora before developing formulations. The aim of this study was to investigate the stability of three progestogens: progesterone, and its two synthetic analogues, medroxyprogesterone acetate (MPA) and levonorgestrel (LNG), in the presence of human colonic microbiota. Progesterone, MPA, and LNG were incubated in mixed fecal inoculum (simulated human colonic fluid) under anerobic conditions. Progesterone was completely degraded after 2 h, whereas levels of MPA and LNG were still detectable after 24 h. The half-lives of progesterone, MPA, and LNG in fecal inoculum were 28, 644, and 240 min, respectively. This study describes the kinetics of colonic microbial metabolism of these hormones for the first time. MPA and LNG show promise for delivery to the colon, potentially improving pharmacokinetics over current oral delivery methods.
Collapse
Affiliation(s)
- Zoe Coombes
- Institute of Life Science 2, Swansea University Medical School, Swansea University, Singleton, Swansea SA28PP, UK;
- Correspondence: (Z.C.); (A.W.B.); (D.G.); Tel.: +44-1792-295384 (Z.C.); +44-1792-602339 (A.W.B.); +44-2077-535865 (D.G.)
| | - Vipul Yadav
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, UK; (V.Y.); (L.E.M.); (C.F.)
| | - Laura E. McCoubrey
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, UK; (V.Y.); (L.E.M.); (C.F.)
| | - Cristina Freire
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, UK; (V.Y.); (L.E.M.); (C.F.)
- Kuecept Limited, Potters Bar, Hertfordshire EN6 1TL, UK
| | - Abdul W. Basit
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, UK; (V.Y.); (L.E.M.); (C.F.)
- Correspondence: (Z.C.); (A.W.B.); (D.G.); Tel.: +44-1792-295384 (Z.C.); +44-1792-602339 (A.W.B.); +44-2077-535865 (D.G.)
| | - R. Steven Conlan
- Institute of Life Science 2, Swansea University Medical School, Swansea University, Singleton, Swansea SA28PP, UK;
| | - Deyarina Gonzalez
- Institute of Life Science 2, Swansea University Medical School, Swansea University, Singleton, Swansea SA28PP, UK;
- Correspondence: (Z.C.); (A.W.B.); (D.G.); Tel.: +44-1792-295384 (Z.C.); +44-1792-602339 (A.W.B.); +44-2077-535865 (D.G.)
| |
Collapse
|
30
|
Zhu Y, Shui X, Liang Z, Huang Z, Qi Y, He Y, Chen C, Luo H, Lei W. Gut microbiota metabolites as integral mediators in cardiovascular diseases (Review). Int J Mol Med 2020; 46:936-948. [PMID: 32705240 PMCID: PMC7388831 DOI: 10.3892/ijmm.2020.4674] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVDs), such as atherosclerosis, hypertension, myocardial infarction and diabetic heart disease, are associated with high morbidity and mortality rates worldwide, and may also induce multiple organ failure in their later stages, greatly reducing the long-term survival of the patients. There are several causes of CVDs, but after nearly a decade of investigation, researchers have found that CVDs are usually accompanied by an imbalance of gut microbiota and a decreased abundance of flora. More importantly, the metabolites produced by intestinal flora, such as trimethylamine and trimethylamine N-oxide, bile acids, short-chain fatty acids and aromatic amino acids, exert different effects on the occurrence and development of CVDs, as observed in the relevant pathways in the cells, which may either promote or protect against CVD occurrence. It is known that changes in the intestinal flora following antibiotic administration, diet supplementation with probiotics, or exercise, can interfere with the composition of the intestinal flora and may represent an effective approach to preventing or treating CVDs. The focus of this review was the analysis of gut microbiota metabolites to elucidate their effects on CVDs and to identify the most cost-effective and beneficial methods for treating CVDs with minimal side effects.
Collapse
Affiliation(s)
- Ying Zhu
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Zheng Liang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Zufeng Huang
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Can Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Lei
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
31
|
Yuan Y, Wu X, Hong Y, Zhang X, Wang Z, Yan H. Salidroside ameliorates liver metabonomics in relation to modified gut-liver FXR signaling in furan-induced mice. Food Chem Toxicol 2020; 140:111311. [DOI: 10.1016/j.fct.2020.111311] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/20/2022]
|
32
|
Ahmadi S, Wang S, Nagpal R, Wang B, Jain S, Razazan A, Mishra SP, Zhu X, Wang Z, Kavanagh K, Yadav H. A human-origin probiotic cocktail ameliorates aging-related leaky gut and inflammation via modulating the microbiota/taurine/tight junction axis. JCI Insight 2020; 5:132055. [PMID: 32302292 DOI: 10.1172/jci.insight.132055] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/08/2020] [Indexed: 12/25/2022] Open
Abstract
Inflammation is a major risk factor of morbidity and mortality in older adults. Although its precise etiology is unknown, low-grade inflammation in older adults is commonly associated with increased intestinal epithelial permeability (leaky gut) and abnormal (dysbiotic) gut microbiota. The increasing older population and lack of treatments to reduce aging-related microbiota dysbiosis, leaky gut, and inflammation culminates in a rise in aging-related comorbidities, constituting a significant public health concern. Here, we demonstrate that a human-origin probiotic cocktail containing 5 Lactobacillus and 5 Enterococcus strains isolated from healthy infant gut prevented high-fat diet-induced (HFD-induced) microbiota dysbiosis, leaky gut, inflammation, metabolic dysfunctions, and physical function decline in older mice. Probiotic-modulated gut microbiota primarily reduced leaky gut by increasing tight junctions, which in turn reduced inflammation. Mechanistically, probiotics modulated microbiota in a way to increase bile salt hydrolase activity, which in turn increased taurine abundance in the gut that stimulated tight junctions and suppressed gut leakiness. Furthermore, in Caenorhabditis elegans, taurine increased life span, reduced adiposity and leaky gut, and enhanced physical function. The results suggest that such probiotic therapies could prevent or treat aging-related leaky gut and inflammation in the elderly.
Collapse
Affiliation(s)
- Shokouh Ahmadi
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Shaohua Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ravinder Nagpal
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Bo Wang
- Department of Chemistry, North Carolina A&T State University, Greensboro, North Carolina, USA
| | - Shalini Jain
- Department of Internal Medicine-Endocrinology and Metabolism.,Mouse Metabolic Phenotyping Core
| | - Atefeh Razazan
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sidharth P Mishra
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Xuewei Zhu
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, and
| | - Zhan Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Kylie Kavanagh
- Department of Pathology-Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Biomedical Sciences, University of Tasmania, Hobart, Australia
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, and
| |
Collapse
|
33
|
Xie Y, Hu F, Xiang D, Lu H, Li W, Zhao A, Huang L, Wang R. The metabolic effect of gut microbiota on drugs. Drug Metab Rev 2020; 52:139-156. [PMID: 32116054 DOI: 10.1080/03602532.2020.1718691] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yuan Xie
- School of Pharmacy, Lanzhou University, Lanzhou, China
- Key Laboratory of the Plateau Medicial, The 940 Hospital of Joint Logistics Support, PLA, Lanzhou, China
| | - Fangdi Hu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Dawei Xiang
- Key Laboratory of the Plateau Medicial, The 940 Hospital of Joint Logistics Support, PLA, Lanzhou, China
| | - Hui Lu
- Key Laboratory of the Plateau Medicial, The 940 Hospital of Joint Logistics Support, PLA, Lanzhou, China
| | - Wenbin Li
- Key Laboratory of the Plateau Medicial, The 940 Hospital of Joint Logistics Support, PLA, Lanzhou, China
| | - Anpeng Zhao
- Key Laboratory of the Plateau Medicial, The 940 Hospital of Joint Logistics Support, PLA, Lanzhou, China
| | - Longji Huang
- School of Pharmacy, Lanzhou University, Lanzhou, China
- Key Laboratory of the Plateau Medicial, The 940 Hospital of Joint Logistics Support, PLA, Lanzhou, China
| | - Rong Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
- Key Laboratory of the Plateau Medicial, The 940 Hospital of Joint Logistics Support, PLA, Lanzhou, China
| |
Collapse
|
34
|
Jia ET, Liu ZY, Pan M, Lu JF, Ge QY. Regulation of bile acid metabolism-related signaling pathways by gut microbiota in diseases. J Zhejiang Univ Sci B 2020; 20:781-792. [PMID: 31489798 DOI: 10.1631/jzus.b1900073] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the past decade, there has been increasing attention on the interaction between microbiota and bile acid metabolism. Bile acids are not only involved in the metabolism of nutrients, but are also important in signal transduction for the regulation of host physiological activities. Microbial-regulated bile acid metabolism has been proven to affect many diseases, but there have not been many studies of disease regulation by microbial receptor signaling pathways. This review considers findings of recent research on the core roles of farnesoid X receptor (FXR), G protein-coupled bile acid receptor (TGR5), and vitamin D receptor (VDR) signaling pathways in microbial-host interactions in health and disease. Studying the relationship between these pathways can help us understand the pathogenesis of human diseases, and lead to new solutions for their treatments.
Collapse
Affiliation(s)
- Er-Teng Jia
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhi-Yu Liu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Pan
- School of Medicine, Southeast University, Nanjing 210097, China
| | - Jia-Feng Lu
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215002, China
| | - Qin-Yu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
35
|
Li H, Xi Y, Xin X, Tian H, Hu Y. Salidroside improves high-fat diet-induced non-alcoholic steatohepatitis by regulating the gut microbiota-bile acid-farnesoid X receptor axis. Biomed Pharmacother 2020; 124:109915. [PMID: 31986416 DOI: 10.1016/j.biopha.2020.109915] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Our previous studies found that salidroside can effectively treat non-alcoholic steatohepatitis (NASH). Here, we discuss the mechanism of salidroside in the treatment of NASH with a focus on the gut microbiota-bile acid-farnesoid X receptor axis. METHODS A NASH mouse model was created by providing mice with a high-fat diet (HFD) for 14 weeks. Mice were randomly divided into the HFD group, HFD + salidroside treatment group, and HFD + obeticholic acid treatment group (n = 8 in each group) and were intragastrically administered corresponding drugs for 4 weeks. Hematoxylin-eosin staining was performed to evaluate the histopathological changes associated with the various treatments. In addition, liver triglyceride (TG) content, serum alanine aminotransferase (ALT) activity, serum inflammatory factors, gut microbiota diversity, and the bile acid profile were evaluated. Western blotting and RT-PCR were performed to detect the expressions of FXR and fibroblast growth factor 15 (FGF15). RESULTS The HFD group displayed obvious signs of hepatic steatosis. The liver TG, serum ALT, and IL-1a, IL-12, MCP-1, KC, MIP-1a, and MIP-1β were significantly higher in the HFD group than the control group (P < 0.01). Intestinal bacteria and bile acid profiles changed significantly in the HFD group (P < 0.05). Further, the expressions of FXR and FGF15 decreased significantly in the HFD group (P < 0.05). After treatment with salidroside, liver steatosis, TG content, and serum inflammatory factors significantly improved and HFD-induced intestinal bacteria, bile acid disorder, and FXR deficiency were significantly alleviated (P < 0.05). CONCLUSION Salidroside can improve NASH via the gut microbiota-bile acid-FXR axis.
Collapse
Affiliation(s)
- Hongshan Li
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Liver Disease Department, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315010, China; Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, 315010, China.
| | - Yingfei Xi
- Medical School of Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Xin Xin
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huajie Tian
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiyang Hu
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
36
|
Liu Y, Wang R, Zheng K, Xin Y, Jia S, Zhao X. Metabonomics analysis of liver in rats administered with chronic low-dose acrylamide. Xenobiotica 2020; 50:894-905. [PMID: 31928121 DOI: 10.1080/00498254.2020.1714791] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current study aimed to investigate the hepatotoxicity of rats administered with chronic low-dose acrylamide (AA) by using metabonomics technology on the basis of ultraperformance liquid chromatography-mass spectrometry (UPLC-MS). A total of 40 male Wistar rats were randomly divided into the following four groups: control, low-dose AA (0.2 mg/kg bw, non-carcinogenic end-point based on the induction of morphological nerve changes in rats), middle-dose AA (1 mg/kg bw), and high-dose AA (5 mg/kg bw). The rats continuously received AA by administering it in drinking water daily for 16 weeks. After the treatment, rat livers were collected for metabonomics analysis and histopathology examination. Principal components analysis (PCA) and partial least-squares discriminant analysis (PLS-DA) were used to investigate the metabonomics profile changes in rat liver tissues and screen the potential biomarkers.Fourteen metabolites were identified with significant changes in intensities (increased or decreased compared with the control group) as a result of treatment (p < 0.05 or p < 0.01). These metabolites included tauro-b-muricholic acid, docosapentaenoic acid, sphingosine 1-phosphate, taurodeoxycholic acid, lysoPE(20:5), cervonyl carnitine, linoleyl carnitine, docosahexaenoic acid, lysoPC(20:4), lysoPE(18:3), PA(20:4), stearidonyl carnitine, alpha-linolenic acid, and lysoPA(18:0).Results showed that chronic exposure to AA at NOAEL (0.2 mg/kg bw) exhibited no toxic effect in rat livers at the metabolic level. AA induced oxidative stress to the liver and disrupted lipid metabolism. The results of liver histopathology examination further supported the metabonomic results.
Collapse
Affiliation(s)
- Yanli Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Ruijuan Wang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Kai Zheng
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Youwei Xin
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Siqi Jia
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Xiujuan Zhao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Kim D, Park C, Meghani NM, Tran TTD, Tran PHL, Park JB, Lee BJ. Utilization of a fattigation platform gelatin-oleic acid sodium salt conjugate as a novel solubilizing adjuvant for poorly water-soluble drugs via self-assembly and nanonization. Int J Pharm 2019; 575:118892. [PMID: 31786354 DOI: 10.1016/j.ijpharm.2019.118892] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/11/2019] [Accepted: 11/16/2019] [Indexed: 12/18/2022]
Abstract
Solubilizing adjuvants are commonly used to dissolve insoluble drugs by simply adding in a formulation. In this study, gelatin and oleic acid sodium salt (OAS), a generally recognized as safe-listed material were chosen and conjugated to develop a natural solubilizing adjuvant using the fattigation platform technology to enhance solubility and dissolution rate of poorly water-soluble drugs according to self-assembly and nanonization principle when simply mixed with poorly water-soluble drugs. We synthesized the gelatin and OAS conjugates (GOC) at three different ratios (1:1, 1:3, 1:5; GOC 1, GOC 2, and GOC 3, respectively) via the 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide/N-hydroxysuccinimide reaction using a spray dryer. This amphiphilic micronized GOC was self-assembled into nanoparticles. The synthesis of new amphiphilic conjugates was identified through Fourier transform-infrared (FT-IR) spectroscopy. The powder properties of the GOCs, such as angle of repose, bulk density, and tapped density were varied with the oleic acid bonding ratio. Then, GOCs were utilized to investigate the enhanced solubility and release rate of various poorly water-soluble drugs such as cilostazol (CSZ), coenzyme Q10, ticagrelor, telmisartan, aprepitant and itraconazole as model drugs. Based on the solubility studies by concentration and type of GOCs, 3% GOC 2 was selected. When this GOC was mixed with these model drugs by the physical mixing, wetting and hot melting methoods, the solubility was highly enhanced compared to the pure control drug, ranging from 20 to 150,000 times. In case of CSZ, all formulations were significantly improved release rate compared to the of CSZ alone and the reference tablet, cilostan® (Korea United Pharm) in simulated intestinal fluid containing 0.2% sodium lauryl sulfate. Differential scanning calorimetry and powder X-ray diffraction were conducted to confirm the crystal polymorphic structure of CSZ, and as a result they changed to diminutive peak intensity compared to CSZ alone. Field-emission scanning electron microscopy indicated that GOC was round with a reduced size of about 100 nm. The reduction of drug particles via nanonization and self-assembly of amphiphilic GOC in an aqueous media could be a key factor to improve poor water solubility by providing a favorable dispersion of drug molecules in an amphiphilic network.
Collapse
Affiliation(s)
- Dayoung Kim
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Chulhun Park
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | | | - Thao T D Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
| | - Phuong H L Tran
- Deakin University, Geelong Australia, School of Medicine, Australia
| | - Jun-Bom Park
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Beom-Jin Lee
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
38
|
Ganci M, Suleyman E, Butt H, Ball M. The role of the brain-gut-microbiota axis in psychology: The importance of considering gut microbiota in the development, perpetuation, and treatment of psychological disorders. Brain Behav 2019; 9:e01408. [PMID: 31568686 PMCID: PMC6851798 DOI: 10.1002/brb3.1408] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The prevalence of psychological disorders remains stable despite steady increases in pharmacological treatments suggesting the need for auxiliary treatment options. Consideration of the brain-gut-microbiota axis (BGMA) has made inroads into reconceptualizing psychological illness from a more holistic perspective. While our understanding of the precise role of gut microbiota (GM) in psychological illness is in its infancy, it represents an attractive target for novel interventions. METHOD An extensive review of relevant literature was undertaken. RESULTS Gut microbiota are proposed to directly and indirectly influence mood, cognition, and behavior which are key components of mental health. This paper outlines how GM may be implicated in psychological disorders from etiology through to treatment and prevention using the Four P model of case formulation. CONCLUSION Moving forward, integration of GM into the conceptualization and treatment of psychological illness will require the discipline of psychology to undergo a significant paradigm shift. While the importance of the GM in psychological well-being must be respected, it is not proposed to be a panacea, but instead, an additional arm to a multidisciplinary approach to treatment and prevention.
Collapse
Affiliation(s)
- Michael Ganci
- Psychology Department, Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia
| | - Emra Suleyman
- Psychology Department, Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia
| | - Henry Butt
- Bioscreen Yarraville (Aust) Pty Ltd, Melbourne, Vic., Australia.,Melbourne University, Melbourne, Vic., Australia
| | - Michelle Ball
- Psychology Department, Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia
| |
Collapse
|
39
|
Ruiz-Canizales J, Domínguez-Avila JA, Wall-Medrano A, Ayala-Zavala JF, González-Córdova AF, Vallejo-Córdoba B, Salazar-López NJ, González-Aguilar GA. Fiber and phenolic compounds contribution to the hepatoprotective effects of mango diets in rats fed high cholesterol/sodium cholate. Phytother Res 2019; 33:2996-3007. [PMID: 31418509 DOI: 10.1002/ptr.6479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/19/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022]
Abstract
The present study evaluated the contribution of mango fiber (MF) and mango phenolic compounds (MP) to the hepatoprotective effect of freeze-dried mango pulp (FDM) cultivar (cv.) "Ataulfo" diets in high cholesterol/sodium cholate (HCC)-fed rats. Male Wistar rats were fed with a HCC diet for 12 weeks, either untreated, or supplemented with MF, MP, FDM, or a control diet (no HCC; n = 6/group). All mango treatments significantly decreased hepatic cholesterol deposition and altered its fatty acid profile, whereas MF and MP mitigated adipose tissue hypertrophy. MF caused a lower level of proinflammatory cytokines (IL-1α/β, IFN-γ, TNF-α) whereas FDM increased the anti-inflammatory ones (IL-4, 6, 10). Mango treatments increased catalase (CAT) activity and its mRNA expression; superoxide dismutase (SOD) activity was normalized by MF and FDM, but its activity was unrelated to its hepatic mRNA expression. Changes in CAT and SOD mRNA expression were unrelated to altered Nrf2 mRNA expression. Higher hepatic PPARα and LXRα mRNA levels were found in MP and MF. We concluded that MF and MP are highly bioactive, according to the documented hepatoprotection in HCC-fed rats; their mechanism of action appears to be related to modulating cholesterol and fatty acid metabolism as well as to stimulating the endogenous antioxidant system.
Collapse
Affiliation(s)
- Jacqueline Ruiz-Canizales
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | | | - Abraham Wall-Medrano
- Departamento de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, México
| | - J Fernando Ayala-Zavala
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | - Aarón F González-Córdova
- Coordinación de Alimentos de Origen Animal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | - Belinda Vallejo-Córdoba
- Coordinación de Alimentos de Origen Animal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | | | - Gustavo A González-Aguilar
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| |
Collapse
|
40
|
Wang XP, Im SJ, Balchak DM, Montalbetti N, Carattino MD, Ray EC, Kashlan OB. Murine epithelial sodium (Na +) channel regulation by biliary factors. J Biol Chem 2019; 294:10182-10193. [PMID: 31092599 PMCID: PMC6664190 DOI: 10.1074/jbc.ra119.007394] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/15/2019] [Indexed: 01/01/2023] Open
Abstract
The epithelial sodium channel (ENaC) mediates Na+ transport in several epithelia, including the aldosterone-sensitive distal nephron, distal colon, and biliary epithelium. Numerous factors regulate ENaC activity, including extracellular ligands, post-translational modifications, and membrane-resident lipids. However, ENaC regulation by bile acids and conjugated bilirubin, metabolites that are abundant in the biliary tree and intestinal tract and are sometimes elevated in the urine of individuals with advanced liver disease, remains poorly understood. Here, using a Xenopus oocyte-based system to express and functionally study ENaC, we found that, depending on the bile acid used, bile acids both activate and inhibit mouse ENaC. Whether bile acids were activating or inhibiting was contingent on the position and orientation of specific bile acid moieties. For example, a hydroxyl group at the 12-position and facing the hydrophilic side (12α-OH) was activating. Taurine-conjugated bile acids, which have reduced membrane permeability, affected ENaC activity more strongly than did their more membrane-permeant unconjugated counterparts, suggesting that bile acids regulate ENaC extracellularly. Bile acid-dependent activation was enhanced by amino acid substitutions in ENaC that depress open probability and was precluded by proteolytic cleavage that increases open probability, consistent with an effect of bile acids on ENaC open probability. Bile acids also regulated ENaC in a cortical collecting duct cell line, mirroring the results in Xenopus oocytes. We also show that bilirubin conjugates activate ENaC. These results indicate that ENaC responds to compounds abundant in bile and that their ability to regulate this channel depends on the presence of specific functional groups.
Collapse
Affiliation(s)
- Xue-Ping Wang
- From the Renal-Electrolyte Division, Department of Medicine
| | | | | | | | - Marcelo D Carattino
- From the Renal-Electrolyte Division, Department of Medicine
- the Department of Cell Biology and Molecular Physiology, and
| | - Evan C Ray
- From the Renal-Electrolyte Division, Department of Medicine
| | - Ossama B Kashlan
- From the Renal-Electrolyte Division, Department of Medicine,
- the Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
41
|
Metformin and Dipeptidyl Peptidase-4 Inhibitor Differentially Modulate the Intestinal Microbiota and Plasma Metabolome of Metabolically Dysfunctional Mice. Can J Diabetes 2019; 44:146-155.e2. [PMID: 31445961 DOI: 10.1016/j.jcjd.2019.05.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/31/2019] [Accepted: 05/22/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Recent evidence indicates that gut microbiota is altered considerably by a variety of commonly prescribed medications. This study assessed the impact of 2 antidiabetic therapeutics on gut microbiota and markers of cardiometabolic disease in metabolically dysfunctional mice. METHODS C57BL/6 mice were fed a high-fat diet for 24 weeks while receiving 1 of 2 antidiabetic therapeutics-metformin or dipeptidyl peptidase-4 (DPP-4) inhibitor, PKF-275-055-for the final 12 weeks. Mice were assessed for weight gain, glucose and cholesterol metabolism, and adiposity. In addition, cecal microbiota was analyzed by 16S compositional sequencing, and plasma metabolome was analyzed by liquid chromatography with tandem mass spectrometry. RESULTS Both therapeutics had similar metabolic effects, attenuating mesenteric adiposity and improving cholesterol metabolism and insulin sensitivity. However, multivariate analyses of microbiota and metabolomics data revealed clear divergence of the therapeutic groups. Although both metformin and PKF-275-055 mice displayed significantly decreased Firmicutes/Bacteroidetes ratios, only metformin harboured metabolic health-associated Akkermansia, Parabacteroides and Christensenella. Paradoxically, metformin also reduced α diversity, a metric frequently associated with host metabolic fitness. PKF-275-055 mice displayed elevated levels of butyrate-producing Ruminococcus and acetogen Dorea, with reduced levels of certain plasma sphingomyelin, phosphatidylcholine and lysophosphatidylcholine entities. In turn, metformin reduced levels of acylcarnitines, a functional group associated with systemic metabolic dysfunction. Finally, several associations were identified between metabolites and altered taxa. CONCLUSIONS This study represents the first direct comparison of the microbiota-modifying effects of metformin and a DPP-4 inhibitor, and proposes several putative microbial targets both in terms of novel therapeutic development and adverse effect prevention.
Collapse
|
42
|
The mechanisms of pharmacokinetic food-drug interactions - A perspective from the UNGAP group. Eur J Pharm Sci 2019; 134:31-59. [PMID: 30974173 DOI: 10.1016/j.ejps.2019.04.003] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/12/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
Abstract
The simultaneous intake of food and drugs can have a strong impact on drug release, absorption, distribution, metabolism and/or elimination and consequently, on the efficacy and safety of pharmacotherapy. As such, food-drug interactions are one of the main challenges in oral drug administration. Whereas pharmacokinetic (PK) food-drug interactions can have a variety of causes, pharmacodynamic (PD) food-drug interactions occur due to specific pharmacological interactions between a drug and particular drinks or food. In recent years, extensive efforts were made to elucidate the mechanisms that drive pharmacokinetic food-drug interactions. Their occurrence depends mainly on the properties of the drug substance, the formulation and a multitude of physiological factors. Every intake of food or drink changes the physiological conditions in the human gastrointestinal tract. Therefore, a precise understanding of how different foods and drinks affect the processes of drug absorption, distribution, metabolism and/or elimination as well as formulation performance is important in order to be able to predict and avoid such interactions. Furthermore, it must be considered that beverages such as milk, grapefruit juice and alcohol can also lead to specific food-drug interactions. In this regard, the growing use of food supplements and functional food requires urgent attention in oral pharmacotherapy. Recently, a new consortium in Understanding Gastrointestinal Absorption-related Processes (UNGAP) was established through COST, a funding organisation of the European Union supporting translational research across Europe. In this review of the UNGAP Working group "Food-Drug Interface", the different mechanisms that can lead to pharmacokinetic food-drug interactions are discussed and summarised from different expert perspectives.
Collapse
|
43
|
Ghaffarzadegan T, Essén S, Verbrugghe P, Marungruang N, Hållenius FF, Nyman M, Sandahl M. Determination of free and conjugated bile acids in serum of Apoe(-/-) mice fed different lingonberry fractions by UHPLC-MS. Sci Rep 2019; 9:3800. [PMID: 30846721 PMCID: PMC6405994 DOI: 10.1038/s41598-019-40272-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
Bile acids (BAs) are known to be involved in cholesterol metabolism but interactions between the diet, BA profiles, gut microbiota and lipid metabolism have not been extensively explored. In the present study, primary and secondary BAs including their glycine and taurine-conjugated forms were quantified in serum of Apoe−/− mice by protein precipitation followed by reversed phase ultra-high-performance liquid chromatography and QTOF mass spectrometry. The mice were fed different lingonberry fractions (whole, insoluble and soluble) in a high-fat setting or cellulose in a high and low-fat setting. Serum concentrations of BAs in mice fed cellulose were higher with the high-fat diet compared to the low-fat diet (20–70%). Among the lingonberry diets, the diet containing whole lingonberries had the highest concentration of chenodeoxycholic acid (CDCA), ursodeoxycholic acid (UDCA), tauro-ursodeoxycholic acid (T-UDCA), α and ω-muricholic acids (MCA) and tauro-α-MCA (T-α-MCA), and the lowest concentration of tauro-cholic acid (T-CA), deoxycholic acid (DCA) and tauro-deoxycholic acid (T-DCA). The glycine-conjugated BAs were very similar with all diets. CDCA, UDCA and α-MCA correlated positively with Bifidobacterium and Prevotella, and T-UDCA, T-α-MCA and ω-MCA with Bacteroides and Parabacteroides.
Collapse
Affiliation(s)
- Tannaz Ghaffarzadegan
- Food for Health Science Centre, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden. .,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden.
| | - Sofia Essén
- Centre for Analysis and Synthesis, Department of Chemistry, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Phebe Verbrugghe
- Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Nittaya Marungruang
- Food for Health Science Centre, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Frida Fåk Hållenius
- Food for Health Science Centre, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Margareta Nyman
- Food for Health Science Centre, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Margareta Sandahl
- Centre for Analysis and Synthesis, Department of Chemistry, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| |
Collapse
|
44
|
Kang JD, Myers CJ, Harris SC, Kakiyama G, Lee IK, Yun BS, Matsuzaki K, Furukawa M, Min HK, Bajaj JS, Zhou H, Hylemon PB. Bile Acid 7α-Dehydroxylating Gut Bacteria Secrete Antibiotics that Inhibit Clostridium difficile: Role of Secondary Bile Acids. Cell Chem Biol 2019; 26:27-34.e4. [PMID: 30482679 PMCID: PMC6338514 DOI: 10.1016/j.chembiol.2018.10.003] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/28/2018] [Accepted: 09/28/2018] [Indexed: 12/11/2022]
Abstract
Clostridium scindens biotransforms primary bile acids into secondary bile acids, and is correlated with inhibition of Clostridium difficile growth in vivo. The aim of the current study was to determine how C. scindens regulates C. difficile growth in vitro and if these interactions might relate to the regulation of gut microbiome structure in vivo. The bile acid 7α-dehydroxylating gut bacteria, C. scindens and C. sordellii, were found to secrete the tryptophan-derived antibiotics, 1-acetyl-β-carboline and turbomycin A, respectively. Both antibiotics inhibited growth of C. difficile and other gut bacteria. The secondary bile acids, deoxycholic acid and lithocholic acid, but not cholic acid, enhanced the inhibitory activity of these antibiotics. These antibiotics appear to inhibit cell division of C. difficile. The results help explain how endogenously synthesized antibiotics and secondary bile acids may regulate C. difficile growth and the structure of the gut microbiome in health and disease.
Collapse
Affiliation(s)
- Jason D Kang
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; McGuire VA Medical Center, Richmond, VA, USA
| | - Christopher J Myers
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Spencer C Harris
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Genta Kakiyama
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; McGuire VA Medical Center, Richmond, VA, USA
| | - In-Kyoung Lee
- NPChem, Co. and Division of Biotechnology, Chonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| | - Bong-Sik Yun
- NPChem, Co. and Division of Biotechnology, Chonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| | - Keiichi Matsuzaki
- Laboratory of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, Nihon University, Chiba, Japan
| | - Megumi Furukawa
- Laboratory of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, Nihon University, Chiba, Japan
| | - Hae-Ki Min
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jasmohan S Bajaj
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; McGuire VA Medical Center, Richmond, VA, USA
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; McGuire VA Medical Center, Richmond, VA, USA
| | - Phillip B Hylemon
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; McGuire VA Medical Center, Richmond, VA, USA.
| |
Collapse
|
45
|
Xue Y, Ma C, Hanna I, Pan G. Intestinal Transporter-Associated Drug Absorption and Toxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:361-405. [DOI: 10.1007/978-981-13-7647-4_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Avelar Rodriguez D, Peña Vélez R, Toro Monjaraz EM, Ramirez Mayans J, Ryan PM. The Gut Microbiota: A Clinically Impactful Factor in Patient Health and Disease. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s42399-018-0036-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Enright EF, Govindarajan K, Darrer R, MacSharry J, Joyce SA, Gahan CGM. Gut Microbiota-Mediated Bile Acid Transformations Alter the Cellular Response to Multidrug Resistant Transporter Substrates in Vitro: Focus on P-glycoprotein. Mol Pharm 2018; 15:5711-5727. [PMID: 30388019 DOI: 10.1021/acs.molpharmaceut.8b00875] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pharmacokinetic research at the host-microbe interface has been primarily directed toward effects on drug metabolism, with fewer investigations considering the absorption process. We previously demonstrated that the transcriptional expression of genes encoding intestinal transporters involved in lipid translocation are altered in germ-free and conventionalized mice possessing distinct bile acid signatures. It was consequently hypothesized that microbial bile acid metabolism, which is the deconjugation and dehydroxylation of the bile acid steroid nucleus by gut bacteria, may impact upon drug transporter expression and/or activity and potentially alter drug disposition. Using a panel of three human intestinal cell lines (Caco-2, T84, and HT-29) that differ in basal transporter expression level, bile acid conjugation-, and hydroxylation-status was shown to influence the transcription of genes encoding several major influx and efflux transporter proteins. We further investigated if these effects on transporter mRNA would translate to altered drug disposition and activity. The results demonstrated that the conjugation and hydroxylation status of the bile acid steroid nucleus can influence the cellular response to multidrug resistance (MDR) substrates, a finding that did not directly correlate with directionality of gene or protein expression. In particular, we noted that the cytotoxicity of cyclosporine A was significantly augmented in the presence of the unconjugated bile acids deoxycholic acid (DCA) and chenodeoxycholic acid (CDCA) in P-gp positive cell lines, as compared to their taurine/glycine-conjugated counterparts, implicating P-gp in the molecular response. Overall this work identifies a novel mechanism by which gut microbial metabolites may influence drug accumulation and suggests a potential role for the microbial bile acid-deconjugating enzyme bile salt hydrolase (BSH) in ameliorating multidrug resistance through the generation of bile acid species with the capacity to access and inhibit P-gp ATPase. The physicochemical property of nonionization is suggested to underpin the preferential ability of unconjugated bile acids to attenuate the efflux of P-gp substrates and to sensitize tumorigenic cells to cytotoxic therapeutics in vitro. This work provides new impetus to investigate whether perturbation of the gut microbiota, and thereby the bile acid component of the intestinal metabolome, could alter drug pharmacokinetics in vivo. These findings may additionally contribute to the development of less toxic P-gp modulators, which could overcome MDR.
Collapse
Affiliation(s)
- Elaine F Enright
- School of Pharmacy , ‡APC Microbiome Ireland , §School of Biochemistry and Cell Biology , ∥School of Microbiology , ⊥School of Medicine , University College Cork , Cork , Ireland
| | | | | | | | | | - Cormac G M Gahan
- School of Pharmacy , ‡APC Microbiome Ireland , §School of Biochemistry and Cell Biology , ∥School of Microbiology , ⊥School of Medicine , University College Cork , Cork , Ireland
| |
Collapse
|
48
|
Pavlović N, Goločorbin-Kon S, Ðanić M, Stanimirov B, Al-Salami H, Stankov K, Mikov M. Bile Acids and Their Derivatives as Potential Modifiers of Drug Release and Pharmacokinetic Profiles. Front Pharmacol 2018; 9:1283. [PMID: 30467479 DOI: 10.3389/fphar.2018.01283/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/18/2018] [Indexed: 05/27/2023] Open
Abstract
Bile acids have received considerable interest in the drug delivery research due to their peculiar physicochemical properties and biocompatibility. The main advantage of bile acids as drug absorption enhancers is their ability to act as both drug solubilizing and permeation-modifying agents. Therefore, bile acids may improve bioavailability of drugs whose absorption-limiting factors include either poor aqueous solubility or low membrane permeability. Besides, bile acids may withstand the gastrointestinal impediments and aid in the transporter-mediated absorption of physically complexed or chemically conjugated drug molecules. These biomolecules may increase the drug bioavailability also at submicellar levels by increasing the solubility and dissolution rate of non-polar drugs or through the partition into the membrane and increase of membrane fluidity and permeability. Most bile acid-induced effects are mediated by the nuclear receptors that activate transcriptional networks, which then affect the expression of a number of target genes, including those for membrane transport proteins, affecting the bioavailability of a number of drugs. Besides micellar solubilization, there are many other types of interactions between bile acids and drug molecules, which can influence the drug transport across the biological membranes. Most common drug-bile salt interaction is ion-pairing and the formed complexes may have either higher or lower polarity compared to the drug molecule itself. Furthermore, the hydroxyl and carboxyl groups of bile acids can be utilized for the covalent conjugation of drugs, which changes their physicochemical and pharmacokinetic properties. Bile acids can be utilized in the formulation of conventional dosage forms, but also of novel micellar, vesicular and polymer-based therapeutic systems. The availability of bile acids, along with their simple derivatization procedures, turn them into attractive building blocks for the design of novel pharmaceutical formulations and systems for the delivery of drugs, biomolecules and vaccines. Although toxic properties of hydrophobic bile acids have been described, their side effects are mostly produced when present in supraphysiological concentrations. Besides, minor structural modifications of natural bile acids may lead to the creation of bile acid derivatives with the reduced toxicity and preserved absorption-enhancing activity.
Collapse
Affiliation(s)
- Nebojša Pavlović
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | | | - Maja Ðanić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Bojan Stanimirov
- Department of Biochemistry, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Hani Al-Salami
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Karmen Stankov
- Department of Biochemistry, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
49
|
Pavlović N, Goločorbin-Kon S, Ðanić M, Stanimirov B, Al-Salami H, Stankov K, Mikov M. Bile Acids and Their Derivatives as Potential Modifiers of Drug Release and Pharmacokinetic Profiles. Front Pharmacol 2018; 9:1283. [PMID: 30467479 PMCID: PMC6237018 DOI: 10.3389/fphar.2018.01283] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/18/2018] [Indexed: 12/16/2022] Open
Abstract
Bile acids have received considerable interest in the drug delivery research due to their peculiar physicochemical properties and biocompatibility. The main advantage of bile acids as drug absorption enhancers is their ability to act as both drug solubilizing and permeation-modifying agents. Therefore, bile acids may improve bioavailability of drugs whose absorption-limiting factors include either poor aqueous solubility or low membrane permeability. Besides, bile acids may withstand the gastrointestinal impediments and aid in the transporter-mediated absorption of physically complexed or chemically conjugated drug molecules. These biomolecules may increase the drug bioavailability also at submicellar levels by increasing the solubility and dissolution rate of non-polar drugs or through the partition into the membrane and increase of membrane fluidity and permeability. Most bile acid-induced effects are mediated by the nuclear receptors that activate transcriptional networks, which then affect the expression of a number of target genes, including those for membrane transport proteins, affecting the bioavailability of a number of drugs. Besides micellar solubilization, there are many other types of interactions between bile acids and drug molecules, which can influence the drug transport across the biological membranes. Most common drug-bile salt interaction is ion-pairing and the formed complexes may have either higher or lower polarity compared to the drug molecule itself. Furthermore, the hydroxyl and carboxyl groups of bile acids can be utilized for the covalent conjugation of drugs, which changes their physicochemical and pharmacokinetic properties. Bile acids can be utilized in the formulation of conventional dosage forms, but also of novel micellar, vesicular and polymer-based therapeutic systems. The availability of bile acids, along with their simple derivatization procedures, turn them into attractive building blocks for the design of novel pharmaceutical formulations and systems for the delivery of drugs, biomolecules and vaccines. Although toxic properties of hydrophobic bile acids have been described, their side effects are mostly produced when present in supraphysiological concentrations. Besides, minor structural modifications of natural bile acids may lead to the creation of bile acid derivatives with the reduced toxicity and preserved absorption-enhancing activity.
Collapse
Affiliation(s)
- Nebojša Pavlović
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | | | - Maja Ðanić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Bojan Stanimirov
- Department of Biochemistry, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Hani Al-Salami
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Karmen Stankov
- Department of Biochemistry, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
50
|
Wiest J, Saedtler M, Böttcher B, Grüne M, Reggane M, Galli B, Holzgrabe U, Meinel L. Geometrical and Structural Dynamics of Imatinib within Biorelevant Colloids. Mol Pharm 2018; 15:4470-4480. [DOI: 10.1021/acs.molpharmaceut.8b00469] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Johannes Wiest
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, DE-97074 Wuerzburg, Germany
| | - Marco Saedtler
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, DE-97074 Wuerzburg, Germany
| | - Bettina Böttcher
- Department of Biochemistry, Rudolf Virchow Center, University of Wuerzburg, Josef-Schneider-Straße 2, DE-97080 Wuerzburg, Germany
| | - Marvin Grüne
- Institute of Organic Chemistry, University of Wuerzburg, DE-97074 Wuerzburg, Germany
| | - Maude Reggane
- Novartis Pharma AG, Lichtstraße 35, CH-4002 Basel, Switzerland
| | - Bruno Galli
- Novartis Pharma AG, Lichtstraße 35, CH-4002 Basel, Switzerland
| | - Ulrike Holzgrabe
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, DE-97074 Wuerzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, DE-97074 Wuerzburg, Germany
| |
Collapse
|