1
|
Tai YH, Kao CY, Zhang YP, Chiou YJ, Chiu HH, Thuy TTD, Liao HW. An integrated platform for investigating drug-microbial interactions to support pharmacomicrobiomics studies. Talanta 2024; 283:127094. [PMID: 39467441 DOI: 10.1016/j.talanta.2024.127094] [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: 07/30/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Investigation of drug-microbial interactions has gained prominence due to the increasing need to study pharmacomicrobiomics. Previous research has revealed the microbiome's role in drug metabolism, influencing efficacy, bioavailability, and toxicity. Several potential interactions have reported between drugs and microbes, including bioaccumulation, biotransformation, and the influence of drugs on microbial growth. To facilitate the investigation of drug-microbial interactions, in this study, we present an integrated platform and procedure for investigating drug-microbial interactions, focusing on biotransformation, bioaccumulation, metabolomics, exometabolomics, lipidomics, and exolipidomics. To illustrate the feasibility of this platform, we examined the interactions between digoxin and Lactiplantibacillus pentosus (L. pentosus), revealing previously unknown interactions. Although the growth of L. pentosus was unaffected by digoxin, metabolomics, exometabolomics, lipidomics, and exolipidomics analyses revealed digoxin's impact on metabolites and lipids inside and outside L. pentosus. Additionally, we utilized a validated liquid chromatography-mass spectrometry quantification platform to evaluate digoxin biotransformation and bioaccumulation levels by L. pentosus. After accurately quantifying digoxin in the supernatant and pellet, we determined that approximately 8.7 % of digoxin was biotransformed by L. pentosus. Exolipidomics analysis further supported digoxin biotransformation, identifying digoxigenin and its metabolites. These findings elucidate the potential impact of L. pentosus on digoxin metabolism, underscoring the importance of considering microbial interactions in pharmacological research. We anticipate that the integrated platform could assist in more pharmacomicrobiomics studies and uncover unknown drug-microbial interactions.
Collapse
Affiliation(s)
- Yu-Hsien Tai
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Yen Kao
- Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Ping Zhang
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Jing Chiou
- Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Huai-Hsuan Chiu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Tran Thi Dieu Thuy
- Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Wei Liao
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
2
|
Mu YF, Gao ZX, Mao ZH, Pan SK, Liu DW, Liu ZS, Wu P. Perspectives on the involvement of the gut microbiota in salt-sensitive hypertension. Hypertens Res 2024; 47:2351-2362. [PMID: 38877311 DOI: 10.1038/s41440-024-01747-y] [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: 11/13/2023] [Revised: 03/13/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024]
Abstract
Salt-sensitivity hypertension (SSH) is an independent predictor of cardiovascular event-related death. Despite the extensiveness of research on hypertension, which covers areas such as the sympathetic nervous system, the renin-angiotensin system, the vascular system, and the immune system, its pathogenesis remains elusive, with sub-optimal blood pressure control in patients. The gut microbiota is an important component of nutritional support and constitutes a barrier in the host. Long-term high salt intake can lead to gut microbiota dysbiosis and cause significant changes in the expression of gut microbiota-related metabolites. Of these metabolites, short chain fatty acids (SCFAs), trimethylamine oxide, amino acids, bile acids, and lipopolysaccharide are essential mediators of microbe-host crosstalk. These metabolites may contribute to the incidence and development of SSH via inflammatory, immune, vascular, and nervous pathways, among others. In addition, recent studies, including those on the histone deacetylase inhibitory mechanism of SCFAs and the blood pressure-decreasing effects of H2S via vascular activation, suggest that several proteins and factors in the classical pathway elicit their effects through multiple non-classical pathways. This review summarizes changes in the gut microbiota and its related metabolites in high-salt environments, as well as corresponding treatment methods for SSH, such as diet management, probiotic and prebiotic use, antibiotic use, and fecal transplantation, to provide new insights and perspectives for understanding SSH pathogenesis and the development of strategies for its treatment.
Collapse
Affiliation(s)
- Ya-Fan Mu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhong-Xiuzi Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zi-Hui Mao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shao-Kang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dong-Wei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| | - Peng Wu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| |
Collapse
|
3
|
Cardoso AM. Microbial influence on blood pressure: unraveling the complex relationship for health insights. MICROBIOME RESEARCH REPORTS 2024; 3:22. [PMID: 38841410 PMCID: PMC11149090 DOI: 10.20517/mrr.2023.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/29/2024] [Accepted: 03/13/2024] [Indexed: 06/07/2024]
Abstract
Hypertension, a critical global health concern, is characterized by persistent high blood pressure and is a major cause of cardiovascular events. This perspective explores the multifaceted implications of hypertension, its association with cardiovascular diseases, and the emerging role of the gut microbiota. The gut microbiota, a dynamic community in the gastrointestinal tract, plays a pivotal role in hypertension by influencing blood pressure through the generation of antioxidant, anti-inflammatory, and short-chain fatty acids metabolites, and the conversion of nitrates into nitric oxide. Antihypertensive medications interact with the gut microbiota, impacting drug pharmacokinetics and efficacy. Prebiotics and probiotics present promising avenues for hypertension management, with prebiotics modulating blood pressure through lipid and cholesterol modulation, and probiotics exhibiting a general beneficial effect. Personalized choices based on individual factors are crucial for optimizing prebiotic and probiotic interventions. In conclusion, the gut microbiota's intricate influence on blood pressure regulation offers innovative perspectives in hypertension therapeutics, with targeted strategies proving valuable for holistic blood pressure management and health promotion.
Collapse
|
4
|
Liu X, Fang H, Pan L, Zhang P, Lin H, Gao H, Ye C, Mao D, Luo Y. S-amlodipine induces liver inflammation and dysfunction through the alteration of intestinal microbiome in a rat model. Gut Microbes 2024; 16:2316923. [PMID: 38400721 PMCID: PMC10896145 DOI: 10.1080/19490976.2024.2316923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 02/06/2024] [Indexed: 02/26/2024] Open
Abstract
S-amlodipine, a commonly prescribed antihypertensive agent, is widely used in clinical settings to treat hypertension. However, the potential adverse effects of long-term S-amlodipine treatment on the liver remain uncertain, given the cautionary recommendations from clinicians regarding its administration in individuals with impaired liver function. To address this, we conducted a study using an eight-week-old male rat model and administered a daily dose of 0.6 ~ 5 mg/kg of S-amlodipine for 7 weeks. Our findings demonstrated that 1.2 ~ 5 mg/kg of S-amlodipine treatment induced liver inflammation and associated dysfunction in rats, further in vitro experiments revealed that the observed liver inflammation and dysfunction were not attributable to direct effects of S-amlodipine on the liver. Metagenome sequencing analysis revealed that S-amlodipine treatment led to alterations in the gut microbiome of rats, with the bloom of E. coli (4.5 ~ 6.6-fold increase) and a decrease in A. muciniphila (1,613.4 ~ 2,000-fold decrease) and B. uniformis (20.6 ~ 202.7-fold decrease), subsequently causing an increase in the gut bacterial lipopolysaccharide (LPS) content (1.4 ~ 1.5-fold increase in feces). S-amlodipine treatment also induced damage to the intestinal barrier and increased intestinal permeability, as confirmed by elevated levels of fecal albumin; furthermore, the flux of gut bacterial LPS into the bloodstream through the portal vein resulted in an increase in serum LPS content (3.3 ~ 4-fold increase). LPS induces liver inflammation and subsequent dysfunction in rats by activating the TLR4 pathway. This study is the first to show that S-amlodipine induces liver inflammation and dysfunction by perturbing the rat gut microbiome. These results indicate the adverse effects of S-amlodipine on the liver and provide a rich understanding of the safety of long-term S-amlodipine administration.
Collapse
Affiliation(s)
- Xinxin Liu
- College of Environmental Sciences and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Hui Fang
- School of Medicine, Nankai University, Tianjin, China
| | - Liuzhu Pan
- School of Medicine, Nankai University, Tianjin, China
| | - Peng Zhang
- College of Environmental Sciences and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Huai Lin
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Huihui Gao
- School of Medicine, Nankai University, Tianjin, China
| | - Chaolin Ye
- School of Medicine, Nankai University, Tianjin, China
| | - Daqing Mao
- School of Medicine, Nankai University, Tianjin, China
| | - Yi Luo
- College of Environmental Sciences and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| |
Collapse
|
5
|
Kim JK, Choi MS, Park HS, Kee KH, Kim DH, Yoo HH. Pharmacokinetic Profiling of Ginsenosides, Rb1, Rd, and Rg3, in Mice with Antibiotic-Induced Gut Microbiota Alterations: Implications for Variability in the Therapeutic Efficacy of Red Ginseng Extracts. Foods 2023; 12:4342. [PMID: 38231867 DOI: 10.3390/foods12234342] [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: 10/25/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024] Open
Abstract
Ginsenoside Rg3 is reported to contribute to the traditionally known diverse effects of red ginseng extracts. Significant individual variations in the therapeutic efficacy of red ginseng extracts have been reported. This study aimed to investigate the effect of amoxicillin on the pharmacokinetics of ginsenosides Rb1, Rd, and Rg3 in mice following the oral administration of red ginseng extracts. We examined the α-diversity and β-diversity of gut microbiota and conducted pharmacokinetic studies to measure systemic exposure to ginsenoside Rg3. We also analyzed the microbiome abundance and microbial metabolic activity involved in the biotransformation of ginsenoside Rb1. Amoxicillin treatment reduced both the α-diversity and β-diversity of the gut microbiota and decreased systemic exposure to ginsenoside Rg3 in mice. The area under the curve (AUC) values for Rg3 in control and amoxicillin-treated groups were 247.7 ± 96.6 ng·h/mL and 139.2 ± 32.9 ng·h/mL, respectively. The microbiome abundance and microbial metabolic activity involved in the biotransformation of ginsenoside Rb1 were also altered by amoxicillin treatment. The metabolizing activity was reduced from 0.13 to 0.05 pmol/min/mg on average. Our findings indicate that amoxicillin treatment potentially reduces the gut-microbiota-mediated metabolism of ginsenoside Rg3 in mice given red ginseng extracts, altering its pharmacokinetics. Gut microbiome variations may thus influence individual ginsenoside pharmacokinetics, impacting red ginseng extract's efficacy. Our results suggest that modulating the microbiome could enhance the efficacy of red ginseng.
Collapse
Affiliation(s)
- Jeon-Kyung Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
- School of Pharmacy, Institute of New Drug Development, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Min Sun Choi
- Pharmacomicrobiomics Research Center, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
| | - Hee-Seo Park
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Kyung Hwa Kee
- Pharmacomicrobiomics Research Center, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
| | - Dong-Hyun Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hye Hyun Yoo
- Pharmacomicrobiomics Research Center, College of Pharmacy, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
6
|
Zhou R, Yang H, Zhu P, Liu Y, Zhang Y, Zhang W, Zhou H, Li X, Li Q. Effect of Gut Microbiota on the Pharmacokinetics of Nifedipine in Spontaneously Hypertensive Rats. Pharmaceutics 2023; 15:2085. [PMID: 37631299 PMCID: PMC10458652 DOI: 10.3390/pharmaceutics15082085] [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: 06/27/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
The pharmacokinetic variability of nifedipine widely observed in the clinic cannot be fully explained by pharmacogenomics. As a new factor affecting drug metabolism, how the gut microbiota affects the pharmacokinetics of nifedipine needs to be explored. Spontaneously hypertensive rats (SHRs) have been commonly used in hypertension-related research and served as the experimental groups; Wistar rats were used as control groups. In this study, the bioavailability of nifedipine decreased by 18.62% (p < 0.05) in the SHRs compared with the Wistar rats. Changes in microbiota were associated with the difference in pharmacokinetics. The relative abundance of Bacteroides dorei was negatively correlated with AUC0-t (r = -0.881, p = 0.004) and Cmax (r = -0.714, p = 0.047). Analysis of serum bile acid (BA) profiles indicated that glycoursodeoxycholic acid (GUDCA) and glycochenodeoxycholic acid (GCDCA) were significantly increased in the SHRs. Compared with the Wistar rats, the expressions of CYP3A1 and PXR were upregulated and the enzyme activity of CYP3A1 increased in the SHRs. Spearman's rank correlation revealed that Bacteroides stercoris was negatively correlated with GUDCA (r = -0.7126, p = 0.0264) and GCDCA (r = -0.6878, p = 0.0339). Moreover, GUDCA was negatively correlated with Cmax (r = -0.556, p = 0.025). In primary rat hepatocytes, GUDCA could induce the expressions of PXR target genes CYP3A1 and Mdr1a. Furthermore, antibiotic treatments in SHRs verified the impact of microbiota on the pharmacokinetics of nifedipine. Generally, gut microbiota affects the pharmacokinetics of nifedipine through microbial biotransformation or by regulating the enzyme activity of CYP3A1.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| | - Haijun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| | - Peng Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| | - Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| | - Yanjuan Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| | - Xiong Li
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510699, China
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| |
Collapse
|
7
|
Bardhan P, Yang T. Sexual Dimorphic Interplays Between Gut Microbiota and Antihypertensive Drugs. Curr Hypertens Rep 2023; 25:163-172. [PMID: 37199902 PMCID: PMC10193343 DOI: 10.1007/s11906-023-01244-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 05/19/2023]
Abstract
PURPOSE OF THE REVIEW The purpose of this study is to review the current literature regarding gut microbiota in blood pressure regulation and its interactions with antihypertensive drugs and to discuss how sex differences in gut microbiota contribute to sexual dimorphism of hypertension and treatment. RECENT FINDINGS The significance of gut microbiota in blood pressure regulation and hypertension etiology is growingly recognized. Targeting the dysbiotic microbiota is proposed to be a new therapeutic method. Recently, a few studies demonstrated that the gut microbiota is highly involved in the modulation of the efficacy of antihypertensive drugs, suggesting a novel mechanism by which gut microbiota plays a role in treatment-resistant hypertension. Furthermore, studies on sex differences in gut microbiota, etiology of hypertension, and sex bias in prescription of antihypertensive medications have revealed promising avenues in sexual dimorphism-based precision medicine. However, no scientific questions are ever raised on how sex differences in gut microbiota contribute to the sex specific responses of certain classes of antihypertensive drugs. Given the dynamics and complexity among individuals, precision medicine is proposed of great potential. We review current knowledge on the interactions between gut microbiota, hypertension, and antihypertensive drugs with an emphasis on sex as a crucial determinant. We propose that sex differences in gut microbiota be a research focus to advance our understanding of hypertension management.
Collapse
Affiliation(s)
- Pritam Bardhan
- Department of Physiology and Pharmacology, Center for Hypertension and Precision Medicine, College of Medicine and Life Sciences, The University of Toledo, Health Science Campus Block Health Science Bldg, Room 310, 3000 Arlington Ave., Toledo, OH, 43614, USA
| | - Tao Yang
- Department of Physiology and Pharmacology, Center for Hypertension and Precision Medicine, College of Medicine and Life Sciences, The University of Toledo, Health Science Campus Block Health Science Bldg, Room 310, 3000 Arlington Ave., Toledo, OH, 43614, USA.
| |
Collapse
|
8
|
Lv J, Wang J, Yu Y, Zhao M, Yang W, Liu J, Zhao Y, Yang Y, Wang G, Guo L, Zhao H. Alterations of gut microbiota are associated with blood pressure: a cross-sectional clinical trial in Northwestern China. J Transl Med 2023; 21:429. [PMID: 37391847 PMCID: PMC10311887 DOI: 10.1186/s12967-023-04176-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/30/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND The human gut microbiota (GM) is involved in the pathogenesis of hypertension (HTN), and could be affected by various factors, including sex and geography. However, available data directly linking GM to HTN based on sex differences are limited. METHODS This study investigated the GM characteristics in HTN subjects in Northwestern China, and evaluate the associations of GM with blood pressure levels based on sex differences. A total of 87 HTN subjects and 45 controls were recruited with demographic and clinical characteristics documented. Fecal samples were collected for 16S rRNA gene sequencing and metagenomic sequencing. RESULTS GM diversity was observed higher in females compared to males, and principal coordinate analysis showed an obvious segregation of females and males. Four predominant phyla of fecal GM included Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria. LEfSe analysis indicated that phylum unidentified_Bacteria was enriched in HTN females, while Leuconostocaceae, Weissella and Weissella_cibaria were enriched in control females (P < 0.05). Functionally, ROC analysis revealed that Cellular Processes (0.796, 95% CI 0.620 ~ 0.916), Human Diseases (0.773, 95% CI 0.595 ~ 0.900), Signal transduction (0.806, 95% CI 0.631 ~ 0.922) and Two-component system (0.806, 95% CI 0.631 ~ 0.922) could differentiate HTN females as effective functional classifiers, which were also positively correlated with systolic blood pressure levels. CONCLUSIONS This work provides evidence of fecal GM characteristics in HTN females and males in a northwestern Chinese population, further supporting the notion that GM dysbiosis may participate in the pathogenesis of HTN, and the role of sex differences should be considered. Trial registration Chinese Clinical Trial Registry, ChiCTR1800019191. Registered 30 October 2018 - Retrospectively registered, http://www.chictr.org.cn/ .
Collapse
Affiliation(s)
- Jing Lv
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jihan Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Yan Yu
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Mengyao Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Wenjuan Yang
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Junye Liu
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yanjie Yang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guodong Wang
- Department of Quality Control, Xi'an Mental Health Center, Xi'an, China
| | - Lei Guo
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Heping Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
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: 5] [Impact Index Per Article: 5.0] [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
|
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: 4.0] [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
|
11
|
Nabeh OA. Gut microbiota and cardiac arrhythmia: a pharmacokinetic scope. Egypt Heart J 2022; 74:87. [PMID: 36583819 PMCID: PMC9803803 DOI: 10.1186/s43044-022-00325-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Dealing with cardiac arrhythmia is a difficult challenge. Choosing between different anti-arrhythmic drugs (AADs) while being cautious about the pro-arrhythmic characteristics of some of these drugs and their diverse interaction with other drugs is a real obstacle. MAIN BODY Gut microbiota (GM), in our bodies, are now being considered as a hidden organ which can regulate our immune system, digest complex food, and secrete bioactive compounds. Yet, GM are encountered in the pathophysiology of arrhythmia and can affect the pharmacokinetics of AADs, as well as some anti-thrombotics, resulting in altering their bioavailability, therapeutic function and may predispose to some of their unpleasant adverse effects. CONCLUSIONS Knowledge of the exact role of GM in the pharmacokinetics of these drugs is now essential for better understanding of the art of arrhythmia management. Also, it will help deciding when to consider probiotics as an adjunctive therapy while treating arrhythmia. This should be discovered in the near future.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- grid.7776.10000 0004 0639 9286Department of Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
12
|
Jia H, Yiyun C, Zhiguo W, Yousong S, Min Z, Yifan S, Na Z, Feng J, Yiru F, Daihui P. Associations between gastrointestinal symptoms, medication use, and spontaneous drug discontinuation in patients with major depressive disorder in China. J Affect Disord 2022; 319:462-468. [PMID: 36055529 DOI: 10.1016/j.jad.2022.08.116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/31/2022] [Accepted: 08/26/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND The study was designed to investigate the associations between gastrointestinal (GI) symptoms, medication use, and spontaneous drug discontinuation (SDD) in patients with major depressive disorder (MDD). METHODS This cross-sectional study included 3256 MDD patients from the National Survey on Symptomatology of Depression (NSSD). Differences in the sociodemographic factors, clinical characteristics, medication use, and self-reported reasons for SDD were compared in patients with different frequencies of GI symptoms. A multiple logistic regression analysis was employed to assess the contribution of GI symptoms to the risk of spontaneous drug discontinuation. RESULTS MDD patients with a higher frequency of GI symptoms were prone to have higher proportions of mood stabilizer and benzodiazepine uses (ps for trend < 0.001) but a lower proportion of SNRI use (pfor trend < 0.001). With the increase in GI symptoms, patients were prone to report worries about long-term side effects (pfor trend < 0.001), with the patients stating ineffective treatments (pfor trend = 0.002) and intolerance of adverse drug reactions (pfor trend = 0.022) as the reasons for SDD. Compared with those patients without GI symptoms, all of the MDD patients with GI symptom frequencies of several days (OR = 1.317; 95 % CI: 1.045-1.660), more than half of all days (OR = 1.305; 95 % CI: 1.005-1.695), and nearly every day (OR = 1.820; 95 %: 1.309-2.531) had an increased risk of SDD. CONCLUSION GI symptoms are highly associated with drug discontinuation in MDD patients. These findings may have important implications for clinical treatment options, as well as for drug adherence management, in MDD patients.
Collapse
Affiliation(s)
- Huang Jia
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China
| | - Cai Yiyun
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China; Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, PR China
| | - Wu Zhiguo
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China
| | - Su Yousong
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China
| | - Zhang Min
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China
| | - Shi Yifan
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China
| | - Zhu Na
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China; Shanghai Pudong New Area Mental Health Center, Shanghai 200122, PR China
| | - Jin Feng
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China
| | - Fang Yiru
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China; Clinical Research Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China.
| | - Peng Daihui
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, PR China.
| |
Collapse
|
13
|
Kyoung J, Atluri RR, Yang T. Resistance to Antihypertensive Drugs: Is Gut Microbiota the Missing Link? Hypertension 2022; 79:2138-2147. [PMID: 35862173 DOI: 10.1161/hypertensionaha.122.19826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Microbiota colonization begins at birth and continuously reshapes throughout the course of our lives, resulting in tremendous interindividual heterogeneity. Given that the gut microbiome, similar to the liver, houses many categories of catalytic enzymes, there is significant value in understanding drug-bacteria interactions. The discovery of this link could enhance the therapeutic value of drugs that would otherwise have a limited or perhaps detrimental effect on patients. Resistant hypertension is one such subset of the hypertensive population that poorly responds to antihypertensive medications, resulting in an increased risk for chronic cardiovascular illnesses and its debilitating effects that ultimately have a detrimental impact on patient quality of life. We recently demonstrated that the gut microbiota is involved in the metabolism of antihypertensive drugs and thus contributes to the pathophysiology of resistant hypertension. Due to a lack of knowledge of the mechanisms, novel therapeutic approaches that account for the gut microbiota may allow for better therapeutic outcomes in resistant hypertension. Therefore, the purpose of this review is to summarize our current, albeit limited, understanding of how the gut microbiota may possess particular enzymatic activities that influence the efficacy of antihypertensive drugs.
Collapse
Affiliation(s)
- Jun Kyoung
- Department of Physiology and Pharmacology, UT Microbiome Consortium, Center for Hypertension and Precision Medicine, College of Medicine and Life Sciences, University of Toledo, OH
| | - Rohit R Atluri
- Department of Physiology and Pharmacology, UT Microbiome Consortium, Center for Hypertension and Precision Medicine, College of Medicine and Life Sciences, University of Toledo, OH
| | - Tao Yang
- Department of Physiology and Pharmacology, UT Microbiome Consortium, Center for Hypertension and Precision Medicine, College of Medicine and Life Sciences, University of Toledo, OH
| |
Collapse
|
14
|
Yang T, Mei X, Tackie-Yarboi E, Akere MT, Kyoung J, Mell B, Yeo JY, Cheng X, Zubcevic J, Richards EM, Pepine CJ, Raizada MK, Schiefer IT, Joe B. Identification of a Gut Commensal That Compromises the Blood Pressure-Lowering Effect of Ester Angiotensin-Converting Enzyme Inhibitors. Hypertension 2022; 79:1591-1601. [PMID: 35538603 PMCID: PMC9278702 DOI: 10.1161/hypertensionaha.121.18711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background: Despite the availability of various classes of antihypertensive medications, a large proportion of hypertensive individuals remain resistant to treatments. The reason for what contributes to low efficacy of antihypertensive medications in these individuals is elusive. The knowledge that gut microbiota is involved in pathophysiology of hypertension and drug metabolism led us to hypothesize that gut microbiota catabolize antihypertensive medications and compromised their blood pressure (BP)-lowering effects. Methods and Results: To test this hypothesis, we examined the BP responses to a representative ACE (angiotensin-converting enzyme) inhibitor quinapril in spontaneously hypertensive rats (SHR) with or without antibiotics. BP-lowering effect of quinapril was more pronounced in the SHR+antibiotics, indicating that gut microbiota of SHR lowered the antihypertensive effect of quinapril. Depletion of gut microbiota in the SHR+antibiotics was associated with decreased gut microbial catabolism of quinapril as well as significant reduction in the bacterial genus Coprococcus. C. comes, an anaerobic species of Coprococcus, harbored esterase activity and catabolized the ester quinapril in vitro. Co-administration of quinapril with C. comes reduced the antihypertensive effect of quinapril in the SHR. Importantly, C. comes selectively reduced the antihypertensive effects of ester ramipril but not nonester lisinopril. Conclusions: Our study revealed a previously unrecognized mechanism by which human commensal C. comes catabolizes ester ACE inhibitors in the gut and lowers its antihypertensive effect.
Collapse
Affiliation(s)
- Tao Yang
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Xue Mei
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Ethel Tackie-Yarboi
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH.,Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH
| | - Millicent Tambari Akere
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH.,Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH
| | - Jun Kyoung
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Blair Mell
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Ji-Youn Yeo
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Xi Cheng
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Elaine M Richards
- Department of Physiology and Functional Genomics (E.M.R., M.K.R.), University of Florida College of Medicine, Gainesville
| | - Carl J Pepine
- Division of Cardiovascular Medicine (C.J.P.), University of Florida College of Medicine, Gainesville
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics (E.M.R., M.K.R.), University of Florida College of Medicine, Gainesville
| | - Isaac T Schiefer
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH.,Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH
| | - Bina Joe
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| |
Collapse
|
15
|
Pant A, Maiti TK, Mahajan D, Das B. Human Gut Microbiota and Drug Metabolism. MICROBIAL ECOLOGY 2022:1-15. [PMID: 35869999 PMCID: PMC9308113 DOI: 10.1007/s00248-022-02081-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 07/18/2022] [Indexed: 05/31/2023]
Abstract
The efficacy of drugs widely varies in individuals, and the gut microbiota plays an important role in this variability. The commensal microbiota living in the human gut encodes several enzymes that chemically modify systemic and orally administered drugs, and such modifications can lead to activation, inactivation, toxification, altered stability, poor bioavailability, and rapid excretion. Our knowledge of the role of the human gut microbiome in therapeutic outcomes continues to evolve. Recent studies suggest the existence of complex interactions between microbial functions and therapeutic drugs across the human body. Therapeutic drugs or xenobiotics can influence the composition of the gut microbiome and the microbial encoded functions. Both these deviations can alter the chemical transformations of the drugs and hence treatment outcomes. In this review, we provide an overview of (i) the genetic ecology of microbially encoded functions linked with xenobiotic degradation; (ii) the effect of drugs on the composition and function of the gut microbiome; and (iii) the importance of the gut microbiota in drug metabolism.
Collapse
Affiliation(s)
- Archana Pant
- Molecular Genetics Lab, National Institute of Immunology, New Delhi, Delhi-110067, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad-121001, India
- Molecular Genetics Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, PO box, Gurgaon Expressway, #04 Faridabad-121001, Haryana, India
| | - Tushar K Maiti
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad-121001, India
| | - Dinesh Mahajan
- Chemistry and Pharmacology Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Bhabatosh Das
- Molecular Genetics Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, PO box, Gurgaon Expressway, #04 Faridabad-121001, Haryana, India.
| |
Collapse
|
16
|
Guo J, Xu Y, Chen LJ, Zhang SX, Liou YL, Chen XP, Tan ZR, Zhou HH, Zhang W, Chen Y. Gut microbiota and host Cyp450s co-contribute to pharmacokinetic variability in mice with non-alcoholic steatohepatitis: Effects vary from drug to drug. J Adv Res 2022; 39:319-332. [PMID: 35777915 PMCID: PMC9263650 DOI: 10.1016/j.jare.2021.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/09/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
Drugs’ pharmacokinetics were changed in NASH disease. A systematical research on cocktail drugs in NASH. Gut microbiota can bio-transform some drugs in vitro, and the metabolic rate was different in NASH. The gut microbiota and the host co-contributed the pharmacokinetic variability of drugs in NASH. The degree of influence on pharmacokinetic variability varies from drug to drug.
Introduction Pharmacokinetic variability in disease state is common in clinical practice, but its underlying mechanism remains unclear. Recently, gut microbiota has been considered to be pharmacokinetically equivalent to the host liver. Although some studies have explored the roles of gut microbiota and host Cyp450s in drug pharmacokinetics, few have explored their effects on pharmacokinetic variability, especially in disease states. Objectives In this study, we aim to investigate the effects of gut microbiota and host Cyp450s on pharmacokinetic variability in mice with non-alcoholic steatohepatitis (NASH), and to elucidate the contribution of gut microbiota and host Cyp450s to pharmacokinetic variability in this setting. Methods The pharmacokinetic variability of mice with NASH was explored under intragastric and intravenous administrations of a cocktail mixture of omeprazole, phenacetin, midazolam, tolbutamide, chlorzoxazone, and metoprolol, after which the results were compared with those obtained from the control group. Thereafter, the pharmacokinetic variabilities of all drugs and their relations to the changes in gut microbiota and host Cyp450s were compared and analyzed. Results The exposures of all drugs, except metoprolol, significantly increased in the NASH group under intragastric administration. However, no significant increase in the exposure of all drugs, except tolbutamide, was observed in the NASH group under intravenous administration. The pharmacokinetic variabilities of phenacetin, midazolam, omeprazole, and chlorzoxazone were mainly associated with decreased elimination activity in the gut microbiota. By contrast, the pharmacokinetic variability of tolbutamide was mainly related to the change in the host Cyp2c65. Notably, gut microbiota and host Cyp450s exerted minimal effects on the pharmacokinetic variability of metoprolol. Conclusion Gut microbiota and host Cyp450s co-contribute to the pharmacokinetic variability in mice with NASH, and the degree of contribution varies from drug to drug. The present findings provide new insights into the explanation of pharmacokinetic variability in disease states.
Collapse
Affiliation(s)
- Jing Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Ying Xu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Li-Jie Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Song-Xia Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Yu-Ligh Liou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Zhi-Rong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China.
| |
Collapse
|
17
|
Ren HC, Sun JG, A JY, Gu SH, Shi J, Shao F, Ai H, Zhang JW, Peng Y, Yan B, Huang Q, Liu LS, Sai Y, Wang GJ, Yang CG. Mechanism-Based Pharmacokinetic Model for the Deglycosylation Kinetics of 20(S)-Ginsenosides Rh2. Front Pharmacol 2022; 13:804377. [PMID: 35694247 PMCID: PMC9175024 DOI: 10.3389/fphar.2022.804377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Aim: The 20(S)-ginsenoside Rh2 (Rh2) is being developed as a new antitumor drug. However, to date, little is known about the kinetics of its deglycosylation metabolite (protopanoxadiol) (PPD) following Rh2 administration. The aim of this work was to 1) simultaneously characterise the pharmacokinetics of Rh2 and PPD following intravenous and oral Rh2 administration, 2) develop and validate a mechanism-based pharmacokinetic model to describe the deglycosylation kinetics and 3) predict the percentage of Rh2 entering the systemic circulation in PPD form. Methods: Plasma samples were collected from rats after the I.V. or P.O. administration of Rh2. The plasma Rh2 and PPD concentrations were determined using HPLC-MS. The transformation from Rh2 to PPD, its absorption, and elimination were integrated into the mechanism based pharmacokinetic model to describe the pharmacokinetics of Rh2 and PPD simultaneously at 10 mg/kg. The concentration data collected following a 20 mg/kg dose of Rh2 was used for model validation. Results: Following Rh2 administration, PPD exhibited high exposure and atypical double peaks. The model described the abnormal kinetics well and was further validated using external data. A total of 11% of the administered Rh2 was predicted to be transformed into PPD and enter the systemic circulation after I.V. administration, and a total of 20% of Rh2 was predicted to be absorbed into the systemic circulation in PPD form after P.O. administration of Rh2. Conclusion: The developed model provides a useful tool to quantitatively study the deglycosylation kinetics of Rh2 and thus, provides a valuable resource for future pharmacokinetic studies of glycosides with similar deglycosylation metabolism.
Collapse
Affiliation(s)
- Hong-can Ren
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- DMPK and Clinical Pharmacology Group, Hutchison MediPharma Ltd., Shanghai, China
- Department of Biology, GenFleet Therapeutics, Shanghai, China
| | - Jian-guo Sun
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ji-ye A
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- *Correspondence: Ji-ye A, ; Guang-ji Wang, ; Cheng-guang Yang,
| | - Sheng-hua Gu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- School of Pharmacy, Shanghai University of Tranditional Chinese Medicine, Shanghai, China
| | - Jian Shi
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Feng Shao
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hua Ai
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jing-wei Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ying Peng
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Bei Yan
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qing Huang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- NMPA Key Laboratory for Impurity Profile of Chemical Drugs, Jiangsu Institute for Food and Drug Control, Nanjing, China
| | - Lin-sheng Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Sai
- DMPK and Clinical Pharmacology Group, Hutchison MediPharma Ltd., Shanghai, China
| | - Guang-ji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- *Correspondence: Ji-ye A, ; Guang-ji Wang, ; Cheng-guang Yang,
| | - Cheng-guang Yang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Ji-ye A, ; Guang-ji Wang, ; Cheng-guang Yang,
| |
Collapse
|
18
|
El Houari A, Ecale F, Mercier A, Crapart S, Laparre J, Soulard B, Ramnath M, Berjeaud JM, Rodier MH, Crépin A. Development of an in vitro Model of Human Gut Microbiota for Screening the Reciprocal Interactions With Antibiotics, Drugs, and Xenobiotics. Front Microbiol 2022; 13:828359. [PMID: 35495704 PMCID: PMC9042397 DOI: 10.3389/fmicb.2022.828359] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Altering the gut microbiota can negatively affect human health. Efforts may be sustained to predict the intended or unintended effects of molecules not naturally produced or expected to be present within the organism on the gut microbiota. Here, culture-dependent and DNA-based approaches were combined to UHPLC-MS/MS analyses in order to investigate the reciprocal interactions between a constructed Human Gut Microbiota Model (HGMM) and molecules including antibiotics, drugs, and xenobiotics. Our HGMM was composed of strains from the five phyla commonly described in human gut microbiota and belonging to Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, and Actinobacteria. Relevantly, the bacterial diversity was conserved in our constructed human gut model through subcultures. Uneven richness distribution was revealed and the sensitivity of the HGMM was mainly affected by antibiotic exposure rather than by drugs or xenobiotics. Interestingly, the constructed model and the individual cultured strains respond with the same sensitivity to the different molecules. UHPLC-MS/MS analyses revealed the disappearance of some native molecules in the supernatants of the HGMM as well as in those of the individual strains. These results suggest that biotransformation of molecules occurred in the presence of our gut microbiota model and the coupled approaches performed on the individual cultures may emphasize new bacterial strains active in these metabolic processes. From this study, the new HGMM appears as a simple, fast, stable, and inexpensive model for screening the reciprocal interactions between the intestinal microbiota and molecules of interest.
Collapse
Affiliation(s)
- Abdelaziz El Houari
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Florine Ecale
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Anne Mercier
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Stéphanie Crapart
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | | | | | | | - Jean-Marc Berjeaud
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| | - Marie-Hélène Rodier
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France.,Laboratoire de Parasitologie et Mycologie, CHU de Poitiers, Poitiers, France
| | - Alexandre Crépin
- UMR CNRS 7267, Laboratoire Ecologie and Biologie des Interactions, Université de Poitiers, Poitiers, France
| |
Collapse
|
19
|
Bai X, Liu G, Yang J, Zhu J, Li X. Gut Microbiota as the Potential Mechanism to Mediate Drug Metabolism Under High-Altitude Hypoxia. Curr Drug Metab 2022; 23:8-20. [PMID: 35088664 DOI: 10.2174/1389200223666220128141038] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/25/2021] [Accepted: 12/30/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The characteristics of pharmacokinetics and the activity and expression of drug-metabolizing enzymes and transporters significantly change under a high-altitude hypoxic environment. Gut microbiota is an important factor affecting the metabolism of drugs through direct or indirect effects, changing the bioavailability, biological activity, or toxicity of drugs and further affecting the efficacy and safety of drugs in vivo. A high-altitude hypoxic environment significantly changes the structure and diversity of gut microbiota, which may play a key role in drug metabolism under a high-altitude hypoxic environment. METHODS An investigation was carried out by reviewing published studies to determine the role of gut microbiota in the regulation of drug-metabolizing enzymes and transporters. Data and information on expression change in gut microbiota, drug-metabolizing enzymes and transporters under a high-altitude hypoxic environment were explored and proposed. RESULTS High-altitude hypoxia is an important environmental factor that can adjust the structure of the gut microbiota and change the diversity of intestinal microbes. It was speculated that the gut microbiota could regulate drug-metabolizing enzymes through two potential mechanisms, the first being through direct regulation of the metabolism of drugs in vivo and the second being indirect, i.e., through the regulation of drug-metabolizing enzymes and transporters, thereby affecting the activity of drugs. CONCLUSION This article reviews the effects of high-altitude hypoxia on the gut microbiota and the effects of these changes on drug metabolism.
Collapse
Affiliation(s)
- Xue Bai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Guiqin Liu
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Jianxin Yang
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
| | - Junbo Zhu
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
| | - Xiangyang Li
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| |
Collapse
|
20
|
Chen HQ, Gong JY, Xing K, Liu MZ, Ren H, Luo JQ. Pharmacomicrobiomics: Exploiting the Drug-Microbiota Interactions in Antihypertensive Treatment. Front Med (Lausanne) 2022; 8:742394. [PMID: 35127738 PMCID: PMC8808336 DOI: 10.3389/fmed.2021.742394] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022] Open
Abstract
Hypertension is a leading risk factor for cardiovascular diseases and can reduce life expectancy. Owing to the widespread use of antihypertensive drugs, patients with hypertension have improved blood pressure control over the past few decades. However, for a considerable part of the population, these drugs still cannot significantly improve their symptoms. In order to explore the reasons behind, pharmacomicrobiomics provide unique insights into the drug treatment of hypertension by investigating the effect of bidirectional interaction between gut microbiota and antihypertensive drugs. This review discusses the relationship between antihypertensive drugs and the gut microbiome, including changes in drug pharmacokinetics and gut microbiota composition. In addition, we highlight how our current knowledge of antihypertensive drug-microbiota interactions to develop gut microbiota-based personalized ways for disease management, including antihypertensive response biomarker, microbial-targeted therapies, probiotics therapy. Ultimately, a better understanding of the impact of pharmacomicrobiomics in the treatment of hypertension will provide important information for guiding rational clinical use and individualized use.
Collapse
Affiliation(s)
- Hui-Qing Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin-Yu Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Kai Xing
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mou-Ze Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Huan Ren
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian-Quan Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- *Correspondence: Jian-Quan Luo
| |
Collapse
|
21
|
Westaway JAF, Huerlimann R, Kandasamy Y, Miller CM, Norton R, Staunton KM, Watson D, Rudd D. The bacterial gut microbiome of probiotic-treated very-preterm infants: changes from admission to discharge. Pediatr Res 2022; 92:142-150. [PMID: 34621029 PMCID: PMC9411061 DOI: 10.1038/s41390-021-01738-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Preterm birth is associated with the development of acute and chronic disease, potentially, through the disruption of normal gut microbiome development. Probiotics may correct for microbial imbalances and mitigate disease risk. Here, we used amplicon sequencing to characterise the gut microbiome of probiotic-treated premature infants. We aimed to identify and understand variation in bacterial gut flora from admission to discharge and in association with clinical variables. METHODS Infants born <32 weeks gestation and <1500 g, and who received probiotic treatment, were recruited in North Queensland Australia. Meconium and faecal samples were collected at admission and discharge. All samples underwent 16S rRNA short amplicon sequencing, and subsequently, a combination of univariate and multivariate analyses. RESULTS 71 admission and 63 discharge samples were collected. Univariate analyses showed significant changes in the gut flora from admission to discharge. Mixed-effects modelling showed significantly lower alpha diversity in infants diagnosed with either sepsis or retinopathy of prematurity (ROP) and those fed formula. In addition, chorioamnionitis, preeclampsia, sepsis, necrotising enterocolitis and ROP were also all associated with the differential abundance of several taxa. CONCLUSIONS The lower microbial diversity seen in infants with diagnosed disorders or formula-fed, as well as differing abundances of several taxa across multiple variables, highlights the role of the microbiome in the development of health and disease. This study supports the need for promoting healthy microbiome development in preterm neonates. IMPACT Low diversity and differing taxonomic abundances in preterm gut microbiota demonstrated in formula-fed infants and those identified with postnatal conditions, as well as differences in taxonomy associated with preeclampsia and chorioamnionitis, reinforcing the association of the microbiome composition changes due to maternal and infant disease. The largest study exploring an association between the preterm infant microbiome and ROP. A novel association between the preterm infant gut microbiome and preeclampsia in a unique cohort of very-premature probiotic-supplemented infants.
Collapse
Affiliation(s)
- Jacob A. F. Westaway
- grid.1011.10000 0004 0474 1797College of Public Health, Medical and Veterinary Science, James Cook University, 1/14-88 McGregor Road, Smithfield, QLD 4878 Australia ,grid.1011.10000 0004 0474 1797Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, 1 James Cook Drive, Douglas, QLD 4811 Australia
| | - Roger Huerlimann
- grid.1011.10000 0004 0474 1797Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, 1 James Cook Drive, Douglas, QLD 4811 Australia ,grid.250464.10000 0000 9805 2626Marine Climate Change Unit, Okinawa Institute of Science and Technology (OIST), 1919-1 Tancha, Onna-son Okinawa, 904-0495 Japan ,grid.1011.10000 0004 0474 1797College of Science and Engineering, James Cook University, 1 James Cook Drive, Douglas, QLD 4811 Australia
| | - Yoga Kandasamy
- grid.1011.10000 0004 0474 1797College of Public Health, Medical and Veterinary Science, James Cook University, 1 James Cook Drive, Douglas, QLD 4811 Australia ,grid.417216.70000 0000 9237 0383Department of neonatology, Townsville University Hospital, 100 Angus Smith Drive, Douglas, QLD 4814 Australia
| | - Catherine M. Miller
- grid.1011.10000 0004 0474 1797College of Public Health, Medical and Veterinary Science, James Cook University, 1/14-88 McGregor Road, Smithfield, QLD 4878 Australia ,grid.1011.10000 0004 0474 1797Australian Institute for Tropical Health and Medicine, James Cook University, 1/14-88 McGregor Road, Smithfield, QLD 4878 Australia
| | - Robert Norton
- Department of Microbiology, Pathology Queensland, 100 Angus Smith Drive, Douglas, QLD 4814 Australia
| | - Kyran M. Staunton
- grid.1011.10000 0004 0474 1797Australian Institute for Tropical Health and Medicine, James Cook University, 1/14-88 McGregor Road, Smithfield, QLD 4878 Australia
| | - David Watson
- grid.417216.70000 0000 9237 0383Department of Maternal-Fetal Medicine, Townsville University Hospital, 100 Angus Smith Drive, Douglas, 4814 Australia
| | - Donna Rudd
- grid.1011.10000 0004 0474 1797College of Public Health, Medical and Veterinary Science, James Cook University, 1 James Cook Drive, Douglas, QLD 4811 Australia
| |
Collapse
|
22
|
Role of the microbiota in hypertension and antihypertensive drug metabolism. Hypertens Res 2021; 45:246-253. [PMID: 34887530 DOI: 10.1038/s41440-021-00804-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/17/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022]
Abstract
Recent evidence suggests that the gut microbiota plays an important role in the development and pathogenesis of hypertension. Dysbiosis, an imbalance in the composition and function of the gut microbiota, was shown to be associated with hypertension in both animal models and humans. In this review, we provide insights into host-microbiota interactions and summarize the evidence supporting the importance of the microbiota in blood pressure (BP) regulation. Metabolites produced by the gut microbiota, especially short-chain fatty acids (SCFAs), modulate BP and vascular responses. Harmful gut-derived metabolites, such as trimethylamine N-oxide and several uremic toxins, exert proatherosclerotic, prothrombotic, and proinflammatory effects. High-salt intake alters the composition of the microbiota, and this microbial alteration contributes to the pathogenesis of salt-sensitive hypertension. In addition, the microbiota may impact the metabolism of drugs and steroid hormones in the host. The drug-metabolizing activities of the microbiota affect the pharmacokinetic parameters of antihypertensive drugs and contribute to the pathogenesis of licorice-induced pseudohyperaldosteronism. Furthermore, the oral microbiota plays a role in BP regulation by producing nitric oxide, which lowers BP via its vasodilatory effects. Thus, antihypertensive intervention strategies targeting the microbiota, such as the use of prebiotics, probiotics, and postbiotics (e.g., SCFAs), are considered new therapeutic options for the treatment of hypertension.
Collapse
|
23
|
Mikrobiota jelitowa a leki. Interakcje wpływające na skuteczność i bezpieczeństwo farmakoterapii. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstrakt
Mikrobiota jelitowa stanowi nieodłączny element organizmu umożliwiający jego prawidłowe funkcjonowanie. Dzięki mikroorganizmom jelitowym możliwa jest stymulacja układu odpornościowego, synteza witamin czy poprawa wchłaniania składników odżywczych. Jednak jej aktywność może również niekorzystnie działać na organizm, m.in. z powodu przetwarzania treści jelitowej. Opisywana w artykule interakcja mikrobiota–lek uwzględnia pozytywny i negatywny wpływ mikroorganizmów jelitowych na farmakoterapię poprzez bezpośrednie i pośrednie oddziaływanie na lek w organizmie. Ze względu na to, że mikrobiom stanowi nieodłączny element organizmu, ingerencja nawet w jego niewielką część może doprowadzić do wystąpienia daleko idących, czasami niespodziewanych skutków. Stąd w celu poprawy skuteczności i bezpieczeństwa farmakoterapii konieczne jest wyjaśnienie mechanizmów oddziaływania mikrobioty na lek w organizmie.
W artykule podsumowano obecną wiedzę na temat biologicznej aktywności mikrobioty jelitowej, a zwłaszcza oddziaływań mikrobiota–leki determinujących skuteczność i bezpieczeństwo farmakoterapii. Wyszukiwanie przeprowadzono we wrześniu 2020 r. w bazach danych PubMed, Scopus, Web of Science, Cochrane Library i powszechnie dostępnej literaturze z użyciem terminów: „mikrobiota jelitowa”, „mikrobiom”, „metabolizm leku”, „interakcje mikrobiota–lek”. W artykule omówiono interakcje między mikrobiotą a lekami m.in. z grupy antybiotyków, inhibitorów pompy protonowej, sulfonamidów, pochodnych kwasu 5-aminosalicylowego, niesteroidowych leków przeciwzapalnych, przeciwnowotworowych, statyn czy metforminą.
Collapse
|
24
|
Kim JK, Choi MS, Kim JY, Yu JS, Seo JI, Yoo HH, Kim DH. Ginkgo biloba leaf extract suppresses intestinal human breast cancer resistance protein expression in mice: Correlation with gut microbiota. Biomed Pharmacother 2021; 140:111712. [PMID: 34010745 DOI: 10.1016/j.biopha.2021.111712] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 02/08/2023] Open
Abstract
In this study, we investigated the effects of treatment with Gingko biloba leaf extract (GLE) on intestinal transporter expression and gut microbiota composition in mice and the correlation between intestinal transporter expression and gut microbiota composition in mice. When GLE was orally administered to mice, intestinal BCRP expression was significantly suppressed. Pharmacokinetic studies showed that the maximum plasma concentration and area under the curve values of sulfasalazine were increased more than twice by treatment with GLE compared with those in the control group. GLE treatment significantly decreased the populations of Proteobacteria and Deferribacteres at the phylum level. Correlation analysis showed that BCRP expression was positively or negatively correlated with the composition of gut bacteria. In Caco-2 cells, GLE treatment did not affect BCRP expression, but treatment with the lysates of GLE-treated mouse feces significantly suppressed BCRP expression. These findings demonstrate that the suppression of intestinal BCRP expression following GLE treatment may occur through modulation of the gut microbiota composition. Thus, the present study suggests that modulation of gut microbiota composition may cause drug transporter-mediated herb-drug interactions.
Collapse
Affiliation(s)
- Jeon-Kyung Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Min Sun Choi
- Institute of Pharmaceutical Science and Technology and College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Jae-Young Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jun Sang Yu
- Institute of Pharmaceutical Science and Technology and College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Jeong In Seo
- Institute of Pharmaceutical Science and Technology and College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hye Hyun Yoo
- Institute of Pharmaceutical Science and Technology and College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea.
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
25
|
Holmes HR, Li Q, Xu K, Kim S, Richards EM, Keeley EC, Handberg EM, Smith SM, Raizada MK, Pepine CJ, Cooper-DeHoff RM. Antihypertensive medication adherence trends by sex and drug class: A pilot study. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2021; 5:100023. [PMID: 38560412 PMCID: PMC10976189 DOI: 10.1016/j.ahjo.2021.100023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/11/2021] [Accepted: 06/10/2021] [Indexed: 04/04/2024]
Abstract
Introduction Antihypertensive medication nonadherence is a prevalent issue but is very difficult to accurately assess. To clarify this problem among hypertensive patients attending a cardiovascular disease outpatient clinic, we utilized high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS) to assess antihypertensive medication adherence and identify trends by sex and drug class. Methods Serum was extracted from blood samples obtained from patients with either drug-controlled or drug resistant hypertension (RHTN) and analyzed via HPLC-MS for antihypertensive drugs which were categorized by drug class as beta blockers, aldosterone antagonists, diuretics, ACE inhibitor/ARBs, or calcium channel blockers. Clinic blood pressure (BP), sex, and prescription regimens were extracted from medical records at or near the time of blood collection. "Adherence" or "nonadherence" was determined by comparison of the patient's prescribed drug regimen and the presence/absence of prescribed drug(s) in their serum. Results Among 76 patients (47 women; mean age 63; 53% white), nonadherence was confirmed in 29%. RHTN was more frequently identified in women than men (55% vs 38%) and nonadherence was higher in women than men (34% vs 21%). BP in those who were adherent to prescribed antihypertensive drugs was significantly lower than in those who were nonadherent (129/75 vs 145/83 mmHg, p = 0.0015). Overall, ACE inhibitors/ARBs were associated with the least nonadherence. Among women, nonadherence was highest for aldosterone antagonists, whereas among men, nonadherence was highest for diuretics. Conclusion We observed nonadherence was more frequent among older women in a cohort of HTN and RHTN patients with cardiovascular disease based on HPLC-MS confirmed drug levels.
Collapse
Affiliation(s)
| | - Qian Li
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ke Xu
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Seungbum Kim
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Elaine M. Richards
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ellen C. Keeley
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Eileen M. Handberg
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Steven M. Smith
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Department of Community Health and Family Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Mohan K. Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rhonda M. Cooper-DeHoff
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
26
|
Balvers M, van den Born BJH, Levin E, Nieuwdorp M. Impact drugs targeting cardiometabolic risk on the gut microbiota. Curr Opin Lipidol 2021; 32:38-54. [PMID: 33332920 DOI: 10.1097/mol.0000000000000727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Alterations in the gut microbiome composition or function are associated with risk factors for cardiometabolic diseases, including hypertension, hyperlipidemia and hyperglycemia. Based on recent evidence that also oral medications used to treat these conditions could alter the gut microbiome composition and function and, vice versa, that the gut microbiome could affect the efficacy of these treatments, we reviewed the literature on these observed interactions. RECENT FINDINGS While the interaction of metformin with the gut microbiome has been studied most, other drugs that target cardiometabolic risk are gaining attention and often showed associations with alterations in microbiome-related features, including alterations in specific microbial taxa or pathways, microbiome composition or microbiome-derived metabolites, while the gut microbiome was also involved in drug metabolism and drug efficacy. As for metformin, for some of them even a potential therapeutic effect via the gut microbiome is postulated. However, exact mechanisms remain to be elucidated. SUMMARY There is growing interest in clarifying the interactions between the gut microbiome and drugs to treat hypertension, hyperlipidemia and hyperglycemia as well as the first pass effect of microbiome on drug efficacy. While mostly analysed in animal models, also human studies are gaining more and more traction. Improving the understanding of the gut microbiome drug interaction can provide clinical directions for therapy by optimizing drug efficacy or providing new targets for drug development.
Collapse
Affiliation(s)
- Manon Balvers
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam
- Horaizon BV, Delft
| | - Bert-Jan H van den Born
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam
- Department of Public Health, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam
- Horaizon BV, Delft
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam
| |
Collapse
|
27
|
Maselis K, Žekevičiūtė R, Vaitkus A. Refractoriness to drugs in migraine may be the result of developing anti-drug antibodies. Med Hypotheses 2020; 146:110459. [PMID: 33360448 DOI: 10.1016/j.mehy.2020.110459] [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: 10/26/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Migraine is a common neurological disease and is listed second among the most disabling health conditions worldwide. Refractory migraine (RM) is a term used to emphasize the unresponsiveness of migraine to various treatment options, encompassing both episodic refractory and chronic refractory migraine. In this paper we discuss various known and possible mechanisms of pharmacological refractoriness in RM, such as possible involvement of the gut microbiome, the blood-brain barrier, migraine genetics and various mechanisms of pharmacokinetic and pharmacodynamic tolerance. Development of medication-overuse headache as a secondary disorder following migraine is also considered. We argue that the available literature is insufficient to fully explain the mechanisms of refractoriness and we present our hypothesis. HYPOTHESIS Refractoriness to drugs in migraine may be the result of developing anti-drug antibodies. Most migraine drugs are small molecules, which cannot elicit an immune response on their own due to their size. However, such molecules can bind to peptide carriers in their vicinity, greatly increasing their immunogenicity. A small molecule possessing this binding ability is called a hapten. Haptens form hapten-carrier complexes (HCCs), which can evoke powerful immune responses. Immune reactions to HCCs are known to be predominantly 'drug allergies' or type 1 drug hypersensitivity reactions', usually resulting from IgE or non-IgE mediated mast cell degranulation. We argue that the immune reaction to HCCs can take shape in developing neutralizing anti-drug antibodies (ADA) in the form of IgG and IgA class antibodies. Since biological therapeutics, such as various monoclonal antibodies, face the issue of ADA-induced drug tolerance, HCCs, being similar in the sense that they carry peptide antigens, are of sufficient size and may be considerably immunogenic, can be responded to in a similar way by producing neutralizing ADA. Furthermore, we argue that such responses are expected to happen more frequently than is thought, due to IgG and IgA being prevalent antibodies, which utilize their neutralizing capabilities on regular basis. Finally, it is important to consider that neutralization reactions in normal immune responses are typically asymptomatic, with the only clinical expression being progressive drug tolerance. These cases may be overshadowed by the life-threatening cases of drug allergy induced anaphylaxis, possibly leading to neutralization reactions being underrecognized. DISCUSSION This hypothesis aims to stimulate more research regarding drug resistance, and if it receives support from empirical evidence, it may help further elucidate the mechanisms underlying refractory diseases and contribute to the development of more effective treatment of many disorders.
Collapse
Affiliation(s)
- K Maselis
- Hospital of Lithuanian, University of Health Sciences Kauno klinikos, Eivenių g. 2, Kaunas, Lithuania.
| | - R Žekevičiūtė
- Hospital of Lithuanian, University of Health Sciences Kauno klinikos, Eivenių g. 2, Kaunas, Lithuania
| | - A Vaitkus
- Lithuanian University of Health Sciences, Eivenių g. 2, Kaunas, Lithuania
| |
Collapse
|
28
|
Koszewicz M, Jaroch J, Brzecka A, Ejma M, Budrewicz S, Mikhaleva LM, Muresanu C, Schield P, Somasundaram SG, Kirkland CE, Avila-Rodriguez M, Aliev G. Dysbiosis is one of the risk factor for stroke and cognitive impairment and potential target for treatment. Pharmacol Res 2020; 164:105277. [PMID: 33166735 DOI: 10.1016/j.phrs.2020.105277] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
More than 50 million people have various forms of cognitive impairment basically caused by neurodegenerative diseases, such as Alzheimer's, Parkinson's, and cerebrovascular diseases as well as stroke. Often these conditions coexist and exacerbate one another. The damaged area in post-stroke dementia may lead to neurodegenerative lesions. Gut microbiome functions like an endocrine organ by generating bioactive metabolites that can directly or indirectly impact human physiology. An alteration in the composition and function of intestinal flora, i.e. gut dysbiosis, is implicated in neurodegenerative and cerebrovascular diseases. Additionally, gut dysbiosis may accelerate the progression of cognitive impairment. Dysbiosis may result from obesity; metabolic disorders, cardiovascular disease, and sleep disorders, Lack of physical activity is associated with dysbiosis as well. These may coexist in various patterns in older people, enhancing the risk, incidence, and progression of cerebrovascular lesions, neurodegenerative disorders, and cognitive impairment, creating a vicious circle. Recently, it has been reported that several metabolites produced by gut microbiota (e.g., trimethylamine/trimethylamine N-oxide, short-chain fatty acids, secondary bile acids) may be linked to neurodegenerative and cerebrovascular diseases. New treatment modalities, including prebiotic and probiotics, may normalize the gut microbiota composition, change the brain-gut barrier, and decrease the risk of the pathology development. Fecal microbiota transplantation, sometimes in combination with other methods, is used for remodeling and replenishing the symbiotic gut microbiome. This promising field of research is associated with basic findings of bidirectional communication between body organs and gut microbiota that creates new possibilities of pharmacological treatments of many clinical conditions. The authors present the role of gut microbiota in physiology, and the novel therapeutic targets in modulation of intestinal microbiota Personalized therapies based on their personal genome make up could offer benefits by modulating microbiota cross-talk with brain and cardiovascular system. A healthy lifestyle, including pre and probiotic nutrition is generally recommended. Prevention may also be enhanced by correcting gut dysbiosis resulting a reduced risk of post-stroke cognitive impairment including dementia.
Collapse
Affiliation(s)
- Magdalena Koszewicz
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Joanna Jaroch
- Faculty of Health Sciences, Wroclaw Medical University, 51-618 Wrocław, Bartla 5, Poland; Department of Cardiology, Lower Silesian Specialist Hospital, Fieldorfa 2, 54-049 Wroclaw, Poland
| | - Anna Brzecka
- Department of Pulmonology and Lung Oncology, Wroclaw Medical University, 53-439, Wroclaw, Grabiszynska 105, Poland
| | - Maria Ejma
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Slawomir Budrewicz
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Liudmila M Mikhaleva
- Federal State Budgetary Institution «Research Institute of Human Morphology», 3, Tsyurupy Str., Moscow, 117418, Russian Federation
| | - Cristian Muresanu
- Research Center for Applied Biotechnology in Diagnosis and Molecular Therapies, Str. Trifoiului nr. 12 G, 400478, Cluj-Napoca, Romania
| | - Pamela Schield
- School of Education & Athletics, Salem University, Salem, WV 26426, United States
| | | | - Cecil E Kirkland
- Department of Biological Sciences, Salem University, Salem, WV, USA
| | - Marco Avila-Rodriguez
- Health Sciences Faculty, Clinic Sciences Department, University of Tolima, 730006 Ibague, Colombia
| | - Gjumrakch Aliev
- Federal State Budgetary Institution «Research Institute of Human Morphology», 3, Tsyurupy Str., Moscow, 117418, Russian Federation; I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432, Russia; GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX, 78229, USA.
| |
Collapse
|
29
|
Tao G, Huang J, Moorthy B, Wang C, Hu M, Gao S, Ghose R. Potential role of drug metabolizing enzymes in chemotherapy-induced gastrointestinal toxicity and hepatotoxicity. Expert Opin Drug Metab Toxicol 2020; 16:1109-1124. [PMID: 32841068 PMCID: PMC8059872 DOI: 10.1080/17425255.2020.1815705] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Toxicity of chemotherapy drugs is the leading cause of poor therapeutic outcome in many cancer patients. Gastrointestinal (GI) toxicity and hepatotoxicity are among the most common side effects of current chemotherapies. Emerging studies indicate that many chemotherapy-induced toxicities are driven by drug metabolism, but very few reviews summarize the role of drug metabolism in chemotherapy-induced GI toxicity and hepatotoxicity. In this review, we highlighted the importance of drug metabolizing enzymes (DMEs) in chemotherapy toxicity. AREAS COVERED Our review demonstrated that altered activity of DMEs play important role in chemotherapy-induced GI toxicity and hepatotoxicity. Besides direct changes in catalytic activities, the transcription of DMEs is also affected by inflammation, cell-signaling pathways, and/or by drugs in cancer patients due to the disease etiology. EXPERT OPINION More studies should focus on how DMEs are altered during chemotherapy treatment, and how such changes affect the metabolism of chemotherapy drug itself. This mutual interaction between chemotherapies and DMEs can lead to excessive exposure of parent drug or toxic metabolites which ultimately cause GI adverse effect.
Collapse
Affiliation(s)
- Gabriel Tao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston TX, U.S
| | - Junqing Huang
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | | | - Cathryn Wang
- Department of Pharmacy Practice and Translational Research, College of Pharmacy, University of Houston, Houston TX, U.S
| | - Ming Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston TX, U.S
| | - Song Gao
- Department of Pharmaceutical and Environmental Health Sciences, Texas Southern University, Houston TX, U.S
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston TX, U.S
| |
Collapse
|
30
|
Yuan T, Wang J, Chen L, Shan J, Di L. Lactobacillus murinus Improved the Bioavailability of Orally Administered Glycyrrhizic Acid in Rats. Front Microbiol 2020; 11:597. [PMID: 32390962 PMCID: PMC7193032 DOI: 10.3389/fmicb.2020.00597] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/18/2020] [Indexed: 12/22/2022] Open
Abstract
Intestinal microbiota has been extensively studied in the context of host health benefit, and it has recently become clear that the gut microbiota influences drug pharmacokinetics and correspondingly efficacy. Intestinal microbiota dysbiosis is closely related with liver cirrhosis, especially the depletion of Lactobacillus. Therefore, the bioavailability of orally administered glycyrrhizic acid (GL) was speculated to be influenced under a pathological state. In the present study, L. murinus was isolated and screened for GL bioconversion capacity in vitro. Compared with Lactobacillus rhamnosus and Lactobacillus acidophilus, L. murinus was chosen for further investigation because it has the highest biotransformation rate. Our results showed that L. murinus could significantly improve the translocation of GL on Caco-2 cell models. Meanwhile, L. murinus was observed to have the ability to bind with the surface of Caco-2 cells and prominently downregulate the transporter gene expression level of multidrug resistance gene 1 (MDR1) and multidrug resistance protein 2 (MRP2), which were involved in the efflux of drugs. Furthermore, L. murinus was selected to be orally administred into rats in healthy and liver cirrhosis groups by a daily gavage protocol. Our data highlighted that supplements of L. murinus significantly improved the bioavailability of orally administered GL in rats, especially under a pathological condition, which may provide a novel strategy for improving the clinical therapeutic effect of liver protective drugs.
Collapse
Affiliation(s)
- Tianjie Yuan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Engineering Research Centre for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Letian Chen
- Jiangsu Engineering Research Centre for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Engineering Research Centre for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
31
|
Li HB, Yang T, Richards EM, Pepine CJ, Raizada MK. Maternal Treatment With Captopril Persistently Alters Gut-Brain Communication and Attenuates Hypertension of Male Offspring. Hypertension 2020; 75:1315-1324. [PMID: 32200676 DOI: 10.1161/hypertensionaha.120.14736] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Maternal-fetal crosstalk has been implicated in long-term control of the health of offspring, including transgenerational hypertension. However, current knowledge is limited regarding maternal influences on the gut and its microbiome in blood pressure control in offspring. Therefore, the current study was designed to test the hypothesis that maternal factors influence the gut-brain axis impacting hypertension in offspring. We elected to use captopril, an antihypertensive angiotensin-converting enzyme inhibitor that possesses antibacterial properties, for the study. Pregnant female spontaneously hypertensive rats and normotensive Wistar Kyoto rats were treated with captopril water (100 mg/[kg·day]) or sterile water throughout pregnancy and lactation. At weaning, the pups from dams drinking sterile water were continued with sterile water until 12 weeks of age. The male pups from dams drinking captopril water were divided at weaning into 2 groups: offspring drinking captopril water and offspring withdrawn from captopril water, then drinking sterile water until 12 weeks of age. Captopril changed gut microbiota of spontaneously hypertensive rat dams, and some of these changes were reflected in their 12-week-old male offspring. These 12-week-old spontaneously hypertensive rat male offspring exposed to captopril via dams demonstrated persistently decreased systolic blood pressure, decreased number of activated microglia and neuroinflammation, as well as improvement of gut inflammation and permeability. Therefore, maternal captopril treatment improves the dysregulated gut-brain axis in spontaneously hypertensive rat male offspring, providing conceptual support that targeting the gut-brain axis via the mother may be a viable strategy for control of hypertension in the offspring.
Collapse
Affiliation(s)
- Hong-Bao Li
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, China (H.-B.L.).,Department of Physiology and Functional Genomics (H.-B.L, T.Y., E.M.R., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Tao Yang
- Department of Physiology and Functional Genomics (H.-B.L, T.Y., E.M.R., M.K.R.), College of Medicine, University of Florida, Gainesville.,Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, OH (T.Y.)
| | - Elaine M Richards
- Department of Physiology and Functional Genomics (H.-B.L, T.Y., E.M.R., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine (C.J.P.), College of Medicine, University of Florida, Gainesville
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics (H.-B.L, T.Y., E.M.R., M.K.R.), College of Medicine, University of Florida, Gainesville
| |
Collapse
|
32
|
Zhang J, Yang J, Yang C, Chen T, Wang Z, Li J, Qin F, Deng Q, Zhang X. Sensitivity to Morphine Reward Associates With Gut Dysbiosis in Rats With Morphine-Induced Conditioned Place Preference. Front Psychiatry 2020; 11:631. [PMID: 33005148 PMCID: PMC7484999 DOI: 10.3389/fpsyt.2020.00631] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022] Open
Abstract
Gut microbiota has been found to establish a bidirectional relationship with the central nervous system. Variations of the gut microbiota has been implicated in various mental disorders, including opioid use disorders. Morphine exposure has been repeatedly found to disrupt the gut microbiota, but association between the gut microbiota and the sensitivity to morphine reward remains unknown. In this study the conditioned place preference (CPP) paradigm was used for morphine-treated rats and saline-treated rats. After the CPP procedure, the morphine-treated rats were divided equally into the low and high CPP (L- and H-CPP) groups according to the CPP scores. We adopted 16S rRNA sequencing for the fecal bacterial communities at baseline and post-conditioning. By comparing the morphine-treated group with saline-treated group, we found alterations of microbial composition in the morphine-treated group, but no significant differences in alpha diversity. The L-CPP group and H-CPP group differed in microbial composition both before and after morphine treatment. The relative abundance of certain taxa was correlated to the CPP scores, such as Alloprevotella and Romboutsia. This study provides direct evidence that morphine exposure alters the composition of the gut microbiota in rats and that microbial alterations are correlated to the sensitivity to morphine reward. These findings may help develop novel therapeutic and preventive strategies for opioid use disorder.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinic Research Center for Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Jun Yang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinic Research Center for Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Cheng Yang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinic Research Center for Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Ti Chen
- Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ziwei Wang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinic Research Center for Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Junyi Li
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinic Research Center for Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Fanglin Qin
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinic Research Center for Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Qijian Deng
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinic Research Center for Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinic Research Center for Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| |
Collapse
|
33
|
Marques FZ, Jama HA, Tsyganov K, Gill PA, Rhys-Jones D, Muralitharan RR, Muir J, Holmes A, Mackay CR. Guidelines for Transparency on Gut Microbiome Studies in Essential and Experimental Hypertension. Hypertension 2019; 74:1279-1293. [DOI: 10.1161/hypertensionaha.119.13079] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Hypertension is a complex and modifiable condition in which environmental factors contribute to both onset and progression. Recent evidence has accumulated for roles of diet and the gut microbiome as environmental factors in blood pressure regulation. However, this is complex because gut microbiomes are a unique feature of each individual reflecting that individual’s developmental and environmental history creating caveats for both experimental models and human studies. Here, we describe guidelines for conducting gut microbiome studies in experimental and clinical hypertension. We provide a complete guide for authors on proper design, analyses, and reporting of gut microbiota/microbiome and metabolite studies and checklists that can be used by reviewers and editors to support robust reporting and interpretation. We discuss factors that modulate the gut microbiota in animal (eg, cohort, controls, diet, developmental age, housing, sex, and models used) and human studies (eg, blood pressure measurement and medication, body mass index, demographic characteristics including age, cultural identification, living structure, sex and socioeconomic environment, and exclusion criteria). We also provide best practice advice on sampling, storage of fecal/cecal samples, DNA extraction, sequencing methods (including metagenomics and 16S rRNA), and computational analyses. Finally, we discuss the measurement of short-chain fatty acids, metabolites produced by the gut microbiota, and interpretation of data. These guidelines should support better transparency, reproducibility, and translation of findings in the field of gut microbiota/microbiome in hypertension and cardiovascular disease.
Collapse
Affiliation(s)
- Francine Z. Marques
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (F.Z.M., H.A.J., K.T., D.R.-J., R.R.M.), Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (F.Z.M., H.A.J.)
| | - Hamdi A. Jama
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (F.Z.M., H.A.J., K.T., D.R.-J., R.R.M.), Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (F.Z.M., H.A.J.)
| | - Kirill Tsyganov
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (F.Z.M., H.A.J., K.T., D.R.-J., R.R.M.), Monash University, Melbourne, Australia
| | - Paul A. Gill
- Translational Nutrition Science in the Department of Gastroenterology, Central Clinical School (P.A.G., J.M., D.R-J.), Monash University, Melbourne, Australia
| | - Dakota Rhys-Jones
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (F.Z.M., H.A.J., K.T., D.R.-J., R.R.M.), Monash University, Melbourne, Australia
| | - Rikeish R. Muralitharan
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (F.Z.M., H.A.J., K.T., D.R.-J., R.R.M.), Monash University, Melbourne, Australia
- Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia (R.R.M.)
| | - Jane Muir
- Translational Nutrition Science in the Department of Gastroenterology, Central Clinical School (P.A.G., J.M., D.R-J.), Monash University, Melbourne, Australia
| | - Andrew Holmes
- Charles Perkin Centre and School of Life and Environmental Sciences, University of Sydney, Australia (A.H.)
| | - Charles R. Mackay
- Infection and Immunity Program, Monash Biomedicine Discovery Institute (C.R.M.), Monash University, Melbourne, Australia
- Department of Biochemistry and Molecular Biology (C.R.M.), Monash University, Melbourne, Australia
| |
Collapse
|
34
|
Iron Transport Tocopheryl Polyethylene Glycol Succinate in Animal Health and Diseases. Molecules 2019; 24:molecules24234289. [PMID: 31775281 PMCID: PMC6930530 DOI: 10.3390/molecules24234289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022] Open
Abstract
Gut health is the starting place for maintaining the overall health of an animal. Strategies to maintain gut health are, thus, an important part in achieving the goal of improving animal health. A new strategy to do this involves two molecules: the iron transport protein ovotransferrin (IT) and α-tocopheryl polyethylene glycol succinate (TPGS), which result in the novel formulation of ITPGS. These molecules help reduce gut pathogens, while enhancing the absorption and bioavailability of therapeutic drugs, phytomedicines, and nanomedicines. This, in turn, helps to maintain normal health in animals. Maintaining the gastrointestinal tract (GIT) in its normal condition is key for successful absorption and efficacy of any nutrient. A compromised GIT, due to an imbalance (dysbiosis) in the GIT microbiome, can lead to an impaired GI barrier system with impaired absorption and overall health of the animal. The molecules in ITPGS may address the issue of poor absorption by keeping the GI system healthy by maintaining the normal microbiome and improving the absorption of nutrients through multiple mechanisms involving antioxidative, anti-inflammatory, immunomodulatory, and antimicrobial activities. The ITPGS technology can allow the dose of active pharmaceutical or herbal medicine to be significantly reduced in order to attain equal or better efficacy. With complimentary actions between IT and TPGS, ITPGS presents a novel approach to increase the bioavailability of drugs, phytoconstituents, nutrients, and nanomedicines by enhanced transport to the tissues at the site of action, while reducing gut pathogen load. The ITPGS approach appears to be a novel strategy for maintaining the health of animals by manipulation of microbiota.
Collapse
|
35
|
Yang J, Li Y, Li Y, Rui X, Du M, Wang Z. Effects of atorvastatin on pharmacokinetics of amlodipine in rats and its potential mechanism. Xenobiotica 2019; 50:685-688. [PMID: 31556335 DOI: 10.1080/00498254.2019.1673918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
| | - Yuanyuan Li
- Department of Imaging, Yidu Central Hospital of Weifang, Weifang, PR China
| | - Yang Li
- Department of Neurosurgery, Zibo No. 4 People’s Hospital, Zibo, PR China
| | - Xueqi Rui
- Department of Cardiovasology, Liyang People’s Hospital, Liyang, PR China
| | - Meiqing Du
- Department of Anesthesiology, Shengli Oilfield Central Hospital, Dongying, PR China
| | - Zengfu Wang
- Department of Anesthesiology, Shengli Oilfield Central Hospital, Dongying, PR China
| |
Collapse
|
36
|
Ðanić M, Stanimirov B, Pavlović N, Vukmirović S, Lazić J, Al-Salami H, Mikov M. Transport and Biotransformation of Gliclazide and the Effect of Deoxycholic Acid in a Probiotic Bacteria Model. Front Pharmacol 2019; 10:1083. [PMID: 31607925 PMCID: PMC6771299 DOI: 10.3389/fphar.2019.01083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/26/2019] [Indexed: 01/04/2023] Open
Abstract
Introduction: Inter-individual differences in gut microflora composition may affect drug metabolism and overall therapeutic response. Gliclazide is a drug characterized by large inter-individual differences in therapeutic response; however, the causes of these differences are not fully explained and may be the outcome of microbial biotransformation. Recently, great attention has been paid to studies on bile acid (BA) interactions with gut microflora and the role of BAs in the modification of drug transport through biological membranes. The Aim: Considering the assumption of gliclazide–probiotic–BAs interactions, the aim of the study was to investigate the transport and biotransformation of gliclazide in probiotic bacteria, as well as the effects of deoxycholic acid (DCA) on gliclazide transport into bacterial cells. Materials and Methods: Probiotics were incubated with gliclazide with or without DCA for 24 h at 37°C. The intracellular and extracellular concentrations of gliclazide were determined at seven time points by high-performance liquid chromatography. Gliclazide biotransformation by the enzymatic activity of probiotic bacteria was examined using appropriate software packages. Results: During the 24 h incubation with probiotic bacteria, significantly lower extracellular concentrations of gliclazide were observed at all time points compared to controls, while in the group with DCA, the decrease in concentration was noticed only at 24 h. The total concentration of gliclazide throughout the whole period was significantly lower compared to control. Proposed pathways of gliclazide biotransformation by probiotic bacteria involve reactions of hydrolysis and hydroxylation. Conclusion: Based on the results obtained, it can be concluded that there are interactions of gliclazide–probiotics–DCA, at both the level of active and passive transport into the cells, and at the level of drug biotransformation by enzymatic activity of probiotic bacteria. The effect of these interactions on the final therapeutic response of gliclazide should be further studied and confirmed in in vivo conditions.
Collapse
Affiliation(s)
- 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
| | - Nebojša Pavlović
- Department of Pharmacy, 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
| | - Jelena Lazić
- Department of Pharmacy, 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
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
37
|
Affiliation(s)
- Hamdi Jama
- From the Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (H.J., D.M.K., F.Z.M.).,Department of Pharmacology (H.J.), Monash University, Melbourne, Australia
| | - David M Kaye
- From the Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (H.J., D.M.K., F.Z.M.).,Central Clinical School (D.M.K.), Monash University, Melbourne, Australia.,Heart Centre, Alfred Hospital, Melbourne, Australia (D.M.K.)
| | - Francine Z Marques
- From the Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (H.J., D.M.K., F.Z.M.).,School of Biological Sciences (F.Z.M.), Monash University, Melbourne, Australia
| |
Collapse
|
38
|
Dam SA, Mostert JC, Szopinska-Tokov JW, Bloemendaal M, Amato M, Arias-Vasquez A. The Role of the Gut-Brain Axis in Attention-Deficit/Hyperactivity Disorder. Gastroenterol Clin North Am 2019; 48:407-431. [PMID: 31383279 DOI: 10.1016/j.gtc.2019.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Genetic and environmental factors play a role in the cause and development of attention-deficit/hyperactivity disorder (ADHD). Recent studies have suggested an important role of the gut-brain axis (GBA) and intestinal microbiota in modulating the risk of ADHD. Here, the authors provide a brief overview of the clinical and biological picture of ADHD and how the GBA could be involved in its cause. They discuss key biological mechanisms involved in the GBA and how these may increase the risk of developing ADHD. Understanding these mechanisms may help to characterize novel treatment options via identification of disease biomarkers.
Collapse
Affiliation(s)
- Sarita A Dam
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Kapittelweg 29, 6525 EN, Nijmegen, The Netherlands.
| | - Jeanette C Mostert
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Joanna W Szopinska-Tokov
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Mirjam Bloemendaal
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Maria Amato
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Alejandro Arias-Vasquez
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands; Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
39
|
Zhang J, Zhang J, Wang R, Jia Z. Effects of Gut Microbiota on Drug Metabolism and Guidance for Rational Drug Use Under Hypoxic Conditions at High Altitudes. Curr Drug Metab 2019; 20:155-165. [PMID: 30338735 DOI: 10.2174/1389200219666181019145159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/11/2018] [Accepted: 09/14/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Modern features of drug development such as low permeability, low solubility, and improved release affect the interplay of the gut microbiota and drug metabolism. In recent years, studies have established the impact of plateau hypoxia on gut microbiota, where drug use by plateau populations is affected by hypoxia- induced changes in intestinal microflora-mediated drug metabolism. METHODS In this review, we summarized the effects of gut microbiota on drug metabolism, and of plateau hypoxia on the intestinal flora, with the aim of providing guidance for the rational use of drugs in high-altitude populations. RESULTS The evidence clearly shows that alterations in gut microbiota can affect pro-drug activation, drug inactivation, and the biotransformation of xenobiotics. Additionally, plateau hypoxia alters drug metabolism by affecting intestinal flora. CONCLUSION This review provides an overview of the effects of gut microbiota on drug metabolism and provides guidance for rational drug use under hypoxic conditions at high altitudes.
Collapse
Affiliation(s)
- Juanhong Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China.,Key Laboratory for Prevention and Remediation of Plateau Environmental Damage, Lanzhou, General Hospital, Lanzhou, 730000, Gansu, China
| | - Junmin Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Rong Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China.,Key Laboratory for Prevention and Remediation of Plateau Environmental Damage, Lanzhou, General Hospital, Lanzhou, 730000, Gansu, China
| | - Zhengping Jia
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China.,Key Laboratory for Prevention and Remediation of Plateau Environmental Damage, Lanzhou, General Hospital, Lanzhou, 730000, Gansu, China
| |
Collapse
|
40
|
Aziz RK, Hegazy SM, Yasser R, Rizkallah MR, ElRakaiby MT. Drug pharmacomicrobiomics and toxicomicrobiomics: from scattered reports to systematic studies of drug-microbiome interactions. Expert Opin Drug Metab Toxicol 2018; 14:1043-1055. [PMID: 30269615 DOI: 10.1080/17425255.2018.1530216] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Pharmacomicrobiomics and toxicomicrobiomics study how variations within the human microbiome (the combination of human-associated microbial communities and their genomes) affect drug disposition, action, and toxicity. These emerging fields, interconnecting microbiology, bioinformatics, systems pharmacology, and toxicology, complement pharmacogenomics and toxicogenomics, expanding the scope of precision medicine. Areas covered: This article reviews some of the most recently reported pharmacomicrobiomic and toxicomicrobiomic interactions. Examples include the impact of the human gut microbiota on cardiovascular drugs, natural products, and chemotherapeutic agents, including immune checkpoint inhibitors. Although the gut microbiota has been the most extensively studied, some key drug-microbiome interactions involve vaginal, intratumoral, and environmental bacteria, and are briefly discussed here. Additionally, computational resources, moving the field from cataloging to predicting interactions, are introduced. Expert opinion: The rapid pace of discovery triggered by the Human Microbiome Project is moving pharmacomicrobiomic research from scattered observations to systematic studies focusing on screening microbiome variants against different drug classes. Better representation of all human populations will improve such studies by avoiding sampling bias, and the integration of multiomic studies with designed experiments will allow establishing causation. In the near future, pharmacomicrobiomic testing is expected to be a key step in screening novel drugs and designing precision therapeutics.
Collapse
Affiliation(s)
- Ramy K Aziz
- a Department of Microbiology and Immunology, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| | - Shaimaa M Hegazy
- b Undergraduate program, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| | - Reem Yasser
- b Undergraduate program, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| | - Mariam R Rizkallah
- c Department of Biometry and Data Management , Leibniz Institute for Prevention Research and Epidemiology - BIPS , Bremen , Germany
| | - Marwa T ElRakaiby
- a Department of Microbiology and Immunology, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| |
Collapse
|