3,084
Views
4
CrossRef citations to date
0
Altmetric
Reviews

The alteration of drug metabolism enzymes and pharmacokinetic parameters in nonalcoholic fatty liver disease: current animal models and clinical practice

, , , , , , & ORCID Icon show all
Pages 163-180 | Received 01 Feb 2023, Accepted 03 Apr 2023, Published online: 28 Apr 2023

Abstract

Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease. The whole concept of NAFLD has now moved into metabolic dysfunction-associated fatty liver disease (MAFLD) to emphasize the strong metabolic derangement as the basis of the disease. Several studies have suggested that hepatic gene expression was altered in NAFLD and NAFLD-related metabolic comorbidities, particularly mRNA and protein expression of phase I and II drug metabolism enzymes (DMEs). NAFLD may affect the pharmacokinetic parameters. However, there were a limited number of pharmacokinetic studies on NAFLD at present. Determining the pharmacokinetic variation in patients with NAFLD remains challenging. Common modalities for modeling NAFLD included: dietary induction, chemical induction, or genetic models. The altered expression of DMEs has been found in rodent and human samples with NAFLD and NAFLD-related metabolic comorbidities. We summarized the pharmacokinetic changes of clozapine (CYP1A2 substrate), caffeine (CYP1A2 substrate), omeprazole (Cyp2c29/CYP2C19 substrate), chlorzoxazone (CYP2E1 substrate), midazolam (Cyp3a11/CYP3A4 substrate) in NAFLD. These results led us to wonder whether current drug dosage recommendations may need to be reevaluated. More objective and rigorous studies are required to confirm these pharmacokinetic changes. We have also summarized the substrates of the DMEs aforementioned. In conclusion, DMEs play an important role in the metabolism of drugs. We hope that future investigations should focus on the effect and alteration of DMEs and pharmacokinetic parameters in this special patient population with NAFLD.

1. Introduction

The biotransformation processes of drugs in vivo are mainly divided into two phases, phase I and phase II drug metabolic reactions. Phase I metabolism consists of functionalization reactions, mainly involving cytochrome P450 (CYP450) enzymes. Phase II drug metabolism is a conjugation reaction that includes glucuronidation, sulfation, and glutathione coupling. The most common phase II drug metabolism enzymes (DMEs) are uridine 5′-diphospho-glucuronosyltransferases (UGTs), glutathione S-transferases (GSTs), and sulfotransferases (SULTs) (Almazroo et al. Citation2017). DMEs are mainly found in the liver, lung, kidney, small intestine, placenta, and skin, but the highest levels are located in the intestinal epithelial cells and the liver (Danielson Citation2002). In chronic liver disease states, the expression and activity of phase I and II DMEs could be altered. The alterations potentially affect the blood/plasma clearance of drugs eliminated by hepatic metabolism or biliary excretion, it can also affect plasma protein binding, which in turn could influence the processes of distribution and elimination (Verbeeck Citation2008).

Nonalcoholic fatty liver disease (NAFLD), the most prevalent chronic liver disease, is defined imageologically or histologically as the presence of hepatic steatosis in absence of secondary hepatic fat accumulation (such as significant alcohol consumption, use of steatogenic medication or hereditary disorders et al.). NAFLD is caused by lipid and peripheral free fatty acids accumulation in the liver. NAFLD is also an umbrella term that encompasses a range from simple hepatic steatosis to nonalcoholic steatohepatitis (NASH). NASH is a more progressive form of NAFLD. A small group of patients can develop NASH, ranging from 3 to 5% (Chalasani et al. Citation2012). NASH is defined as a more severe state of vacuolation, inflammation, and hepatocyte damage (Friedman et al. Citation2018). Generally, NASH is accompanied by pericellular fibrosis and may progress to fibrosis, cirrhosis, or hepatocellular carcinoma (Oseini and Sanyal Citation2017). In 2016, the global NAFLD prevalence was 25%; this increased to >30% in 2019. Prevalence in Asia, Latin America, and Middle East-North Africa was 30.8%, 34.5%, and 42.6%, respectively (Henry et al. Citation2022). In addition, a large proportion of patients with NAFLD have metabolic comorbidities. Metabolic comorbidities encompass obesity (51.34%), type 2 diabetes mellitus (T2DM) (22.51%), hypertension (39.34%), dyslipidemia (69.16%), and metabolic syndrome (42.54%), with insulin resistance as the common underlying pathophysiology (Cobbina and Akhlaghi Citation2017; Sakurai et al. Citation2021). While NAFLD largely increases the risk of cardiovascular disease and extrahepatic cancers. As a result, the burden of NAFLD has become a major public health issue (Zhou, Zhou, et al. Citation2020).

Many studies have reported that metabolic dysfunction is closely involved in the complex mechanism underlying the development of NAFLD, which has prompted a movement to consider renaming NAFLD as metabolic dysfunction-associated fatty liver disease (MAFLD) (Lin et al. Citation2020; Gofton et al. Citation2022). The whole concept of NAFLD has now moved into MAFLD to emphasize the strong metabolic derangement as the basis of the disease. MAFLD is diagnosed based on evidence of hepatic steatosis (detected by imaging, blood biomarkers, or liver histology) and the presence of any one of the following three criteria, namely overweight/obesity, presence of T2DM, or evidence of metabolic dysregulation (Eslam et al. Citation2020). Metabolic dysfunction in this context encompasses obesity, T2DM, hypertension, dyslipidemia, and metabolic syndrome (a collection of clinical conditions including hypertension, atherosclerotic dyslipidemia, obesity, insulin resistance, or T2DM). Whether the variation of DMEs in NAFLD was consisted with those in NAFLD-related metabolic comorbidities remains poorly understood.

Due to the uncertain inner mechanisms of NAFLD, no valid therapy has been approved. Current first-line treatments rely on dietary management, lifestyle modification, and physical exercise. There are many drugs targeting diverse molecular mechanisms of NAFLD in the pipeline, such as dapagliflozin, aramchol, resmetirom, semaglutide, and lanifibranor (Negi et al. Citation2022). Existing drugs such as antidiabetic, anti-obesity, antioxidants, and cytoprotective agents have also been considered in the management of NAFLD and NAFLD-related metabolic comorbidities (Polyzos et al. Citation2020; Negi et al. Citation2022). Several studies found that the expression and activity of DMEs were altered in NAFLD and NAFLD-related metabolic comorbidities (Buechler and Weiss Citation2011; Cobbina and Akhlaghi Citation2017; Wang et al. Citation2020). These alterations could be potential sources of drug variability in patients with NAFLD and could have serious consequences on safety and efficacy, yet it remains unclear. In this review, we summarized the current common animal models of NAFLD and the variation of phase I and II DMEs in NAFLD and NAFLD-related metabolic comorbidities. Meanwhile, we have a specific focus on the pharmacokinetics changes and their impact on clinical outcomes for patients with NAFLD.

2. Animal models of NAFLD

At present, liver biopsy is the gold standard for characterizing liver histology in patients with NAFLD. However, it is expensive and several studies have indicated significant risks and complications associated with liver biopsies, including pain, major bleeding, and death (Chalasani et al. Citation2012). Because collecting liver tissues from NAFLD patients was difficult due to the invasive nature of the technique, animal models (mice or rats) were generally used to analyze gene alterations in NAFLD. Common modalities for modeling NAFLD included: dietary induction, chemical induction, or genetic models (). And most models successfully mimicked the histological pattern of NAFLD.

Table 1. The most commonly used mouse and rat models of NAFLD.

2.1. Dietary induction

Dietary induction models of NAFLD were established by feeding mice or rats in high-fat, sugar, cholesterol, methionine, and choline-deficient diets. There were three types of diet-induced models depending on the feeding component. The first type was defined by reduced lipid export or catabolism ((methionine and choline-deficient diet (MCDD); choline-deficient l-amino acid-defined diet (CDAAD)) (Nakanishi et al. Citation2019; Zhang, Li, et al. Citation2021). The second type included models with increased lipid import or synthesis in the liver (fructose, high-fat emulsion, high-fat diet (HFD)) (Zou et al. Citation2006; Gao et al. Citation2020; Cho et al. Citation2021). The third type was an HFD combined with another diet, including a high-fat high-fructose diet (HFHFD), choline-deficient high-fat diet (CDHFD), high-fat high-sucrose diet (HFHSD), and high-fat high-cholesterol diet (HFHCD) (Yan et al. Citation2018; Baiges-Gaya et al. Citation2021; Hong et al. Citation2022; Lad et al. Citation2022). Hepatic steatosis, hepatic lipid droplet formation, hepatocyte ballooning, inflammatory cell infiltration, liver damage, and liver fibrosis were frequently observed in these models. Dietary induction models of NAFLD have been listed in .

2.2. Combination induction of HFD and chemical

HFD is used to induce not only NAFLD but also metabolic syndrome, dyslipidemia, obesity, and insulin resistance (Zhou, Li, et al. Citation2020; Kumar et al. Citation2021; Zheng, Zhu, et al. Citation2021). However, developing the pathological features of T2DM or NASH is a lengthy process. HFD and treatment with various chemicals affecting hepatic physiology in mice or rats accelerated T2DM, NASH, and fibrosis formation. These chemicals included streptozotocin (STZ), tetrachloride (CCl4), diethylnitrosamine (DEN), and thioacetamide (TH). HFD combing with STZ was used to induce T2DM (Zhang et al. Citation2018; Liu, Deng, et al. Citation2019). CCl4 exacerbated hepatocyte ballooning, inflammation, fibrosis stage, ductular reaction, hepatocyte proliferation, and tumor development when mice were fed a western diet (high-fat, high-fructose, and high-cholesterol) (Tsuchida et al. Citation2018). HFD plus CCl4 could induce the NASH model in vivo (Huang, Wu, et al. Citation2021). A hepatocellular carcinoma model was created using DEN (Stowers et al. Citation1988; Jaworski et al. Citation2005; Connor et al. Citation2018). Animal models of NASH induced by HFD combined with DEN contribute to tumor progression (Sylvester Darvin et al. Citation2018; Márquez-Quiroga et al. Citation2022). TH has been widely used to induce acute or chronic liver injury in mice and rats. Fast food diet (FFD) (high saturated fats, cholesterol, and fructose) combined with TH was used to induce NASH and fibrosis (Sharma et al. Citation2019). The combination induction of HFD and chemicals has been listed in .

2.3. Genetic models

The principal genetic animal models of NAFLD are the mice lacking farnesoid X receptor (Fxr-null mice), apolipoprotein E-deficient (ApoE−/−) mice, leptin-deficient (ob/ob) mice, leptin receptor-deficient (db/db) mice, and Zucker fatty rats (fa/fa, ZFR). In hepatic histology, Fxr-null mice exhibited hepatomegaly, hepatic steatosis, and hepatic inflammation (Miyata, Funaki, et al. Citation2020; Miyata, Matsushita, et al. Citation2020). Unlike Fxr-null mice, ApoE-/- mice developed spontaneous hypercholesterolemia and atherosclerosis and were more susceptible to accelerated liver aging and severe liver damage (Hua et al. Citation2021). The strength of the ob/ob and db/db mice was that the phenotype of these mice mimicked the human metabolic syndrome condition in many ways. However, with congenital leptin deficiency and leptin resistance caused by gene mutations, the weakness of these mice was that they didn’t spontaneously develop steatohepatitis or liver fibrosis (Takahashi et al. Citation2012). In rats, the ZFR was the most commonly used genetic model of NAFLD. The ZFR was widely used as an animal model for genetic obesity, insulin resistance, and metabolic syndrome (Matsumoto et al. Citation2021). Genetic models of NAFLD have been listed in .

3. The alteration of phase I DMEs in NAFLD and metabolic comorbidities

Drug metabolism processes can be classified as phase I and phase II reactions. Phase I reactions, such as oxidation, reduction, and hydrolysis, introduce reactive or polar groups (-OH, -COOH, -NH2, -SH, etc.) into drugs. This process is primarily mediated by the CYP450 enzymes, which are mainly oxidases, reductases, and hydrolases. The CYP450 genes are mainly divided into the CYP1, CYP2, and CYP3 subfamilies and are responsible for the metabolism of approximately 73% of clinical drugs, including CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 (Zhao et al. Citation2021).

Numerous animal studies have observed significant hepatic DMEs changes in NAFLD and metabolic comorbidities. The details and results of these studies have been presented in the following tables ( and ). CYP1A2, a member of the CYP1 family, is exclusively expressed in the human liver (Faber et al. Citation2005). It is responsible for the metabolism of 9% of the drugs used today (Zhao et al. Citation2021), and it can convert some polycyclic aromatic hydrocarbons into carcinogenic intermediates (Vogel et al. Citation2020). CYP2 is the largest family of CYPs, and CYP2D6 and CYP2C19 are responsible for the metabolism of about 27% of the 248 drugs (Zhao et al. Citation2021). Studies have shown that the other isoforms of the CYP2 family have the following rank order in terms of the total hepatic CYP enzyme content: CYP2E1 >CYP2C9>CYP2D6>CYP2A6>CYP2C19>CYP2B6 (Song et al. Citation2021). The majority of NAFLD animal models showed that the expression and activity of CYP1A2, CYP2C19, and CYP2D6 were inhibited. Results from different adipomorphic rat and ob/ob mouse models showed that the mRNA expression, protein level, and activity of Cyp1a2 were significantly down-regulated (Osabe et al. Citation2008; Lee et al. Citation2013; Chang et al. Citation2014; Li, Clarke, et al. Citation2017; Li et al. Citation2021; Wu et al. Citation2023). However, the mRNA expression of Cyp1a2 in C57BL/6 mice fed with the HFHSD or HFD was up-regulated (Chiba et al. Citation2016; Wang et al. Citation2020). The mRNA expression alteration of Cyp1a2 in NAFLD was not entirely consistent between C57BL/6 mice and ob/ob mice. In HFD-induced obese rats, the mRNA expression, protein level, and activity of CYP1A2 were all down-regulated (Zhang et al. Citation2019). The altered expression trend of CYP1A2 in obesity is consistent with those in HFD rats. It was also found that the expression of CYP1A2 decreased in hypertension and hypercholesterolemia rat models (Niu et al. Citation2022; Xu et al. Citation2022). In contrast, the mRNA expression and activity of CYP1A2 were up-regulated in T2DM rats (Wang et al. Citation2018; Yao et al. Citation2020). The activity of CYP1A2 and CYP2C19 decreased in adults with NASH (Fisher et al. Citation2009; Li, Canet, et al. Citation2017). While both the mRNA expression and activity of Cyp2c29/CYP2C19 were all up-regulated in CDAHFD mice (Suzuki et al. Citation2020). And the protein expression and activity of Cyp2c29 (homologues human CYP2C19) (Li, Clarke, et al. Citation2017), Cyp2d22 (homologues human CYP2D6) (Chiba et al. Citation2016) are down-regulated in animal models feeding on HFD or ob/ob mice (Li, Clarke, et al. Citation2017; Wu et al. Citation2023).

Table 2. The alteration of phase I DMEs in NAFLD and metabolic comorbidities.

Table 3. The alteration of phase II DMEs in NAFLD and metabolic comorbidities.

In human and mouse models, the mRNA expression, protein level, and activity of CYP2A6 were all up-regulated (Fisher et al. Citation2009; Li et al. Citation2013; Wang et al. Citation2017; Wang et al. Citation2019). C57BL/6 mice were fed with the HFD for eight weeks, which resulted in significant inhibition of CYP2B6 activity (Wu et al. Citation2023). It was found that both mRNA and protein expression of CYP2C9 were decreased (Li, Clarke, et al. Citation2017). In hypertension rats, the activity of CYP2B6, CYP2D6, and Cyp2c29/CYP2C19 was all decreased (Niu et al. Citation2022). Simultaneously, the same trend was observed in HFD mice. In T2DM, the protein level and activity of Cyp2c29/CYP2C19 were all down-regulated (Yao et al. Citation2019; Kvitne et al. Citation2022). In humans, CYP2B6 and CYP2C9 mRNA expression increased with NAFLD progression, while both the mRNA and protein expression of CYP2E1 tended to decrease with NAFLD progression (Fisher et al. Citation2009). In contrast, the mRNA and protein expression of Cyp2e1 were up-regulated in HFD rats and mice (Zou et al. Citation2006; Li et al. Citation2011; Wang et al. Citation2020; Liu et al. Citation2022). The mRNA expression, protein level, and activity of CYP2E1 were all up-regulated in obesity, dyslipidemia, and metabolic syndrome mice and T2DM rats (Wang et al. Citation2018, Citation2019, Citation2020; Xu et al. Citation2019; Maksymchuk et al. Citation2022). In hypercholesterolemia, the mRNA and protein expression of CYP2E1 was also up-regulated in HFD mice (Wang et al. Citation2020), while both the mRNA expression and activity of CYP2E1 decreased in rats (Xu et al. Citation2022).

CYP3A4 enzyme is responsible for the metabolism of over 50% of the drugs in clinically used drugs and accounts for nearly 22% of the total hepatic CYP enzymes (Song et al. Citation2021). In patients with NAFLD and HFD-fed mice, the mRNA expression, protein level, and activity of CYP3A4 were down-regulated (Wahlang et al. Citation2014; Woolsey et al. Citation2015; Chiba et al. Citation2016; Jamwal et al. Citation2018). The mRNA expression of Cyp3a11 (homologues human CYP3A4) (Chiba et al. Citation2016) was significantly increased in ob/ob mice. However, a significant decrease in protein expression of Cyp3a11/CYP3A4 was observed in ob/ob MCDD-fed mice. In hypertension rats, the activity of Cyp3a11/CYP3A4 was down-regulated (Niu et al. Citation2022). However, the activity of Cyp3a11/CYP3A4 was not consistent in rats with T2DM (Yao et al. Citation2019, Citation2020). The regulation of Phase I DMEs protein expression in NAFLD is the consistent finding. However, the data about mRNA expression and activity of the murine CYP isoforms are not in complete concordance with each other, which may be indicative of the heterogeneity in the regulation mechanisms known for each isoform (Li, Clarke, et al. Citation2017).

4. Effect of NAFLD and metabolic comorbidities on phase II DMEs

Phase II metabolic reactions, also known as binding reactions, are processes in which the products of phase I reactions are coupled to appropriate functional groups to produce water-soluble compounds that contribute to the detoxification of endogenous and exogenous substrates (Pathania et al. Citation2018). UGTs are responsible for the glucuronidation process. These enzymes bind glucuronic acid to small non-polar molecules, producing glucuronides that are usually inactive and water-soluble, thus facilitating their excretion from the body through the urine or feces. At the same time, the activity of the drug is generally reduced. Thus, glucuronidation detoxifies a broad array of exogenous compounds including environmental toxins, carcinogens, and commonly prescribed drugs. The main UGTs involved in the glucuronidation of drugs in the liver, including UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2A3, UGT2B1, and UGT2B7 (Meech et al. Citation2019). In the UGT1 family, UGT1A1 is involved in the glucuronidation of bilirubin (Lévesque et al. Citation2007). Two isozymes, UGT1A4 and UGT1A3, were identified as the principal catalysts of 25-Hydroxyvitamin D3 glucuronidation in the human liver (Wang et al. Citation2014). UGT1A6 has specificity for planar phenols and arylamines (Bock and Köhle Citation2005). UGT1A9 is involved in the biotransformation of 7-ethyl-10-hydroxycamptothecin (Gagné et al. Citation2002). In the UGT2 family, UGT2A3 predominantly glucuronidates hyodeoxycholic acid at the 6-hydroxy position (Court et al. Citation2008). Bisphenol A is glucuronidated by UGT2B1 (Yokota et al. Citation1999). UGT2B7 demonstrated the highest catalytic activities for estrogens (Lépine et al. Citation2004). In steatotic livers of ob/ob mice, Ugt1a1, Ugt1a6, Ugt1a9, and Ugt2a3 mRNA expression increased (Xu et al. Citation2012). In male C57BL/6 mice fed with the HFD, the mRNA expression of Ugt1a1 was higher than those with a low-fat diet at 8 and 18 weeks (Wang et al. Citation2020). In the HFHSD rats, consistent with the mRNA expression, the protein expression of Ugt1a1, Ugt1a6, Ugt1a7, and Ugt2b1 were significantly increased, whereas the protein levels of Ugt1a1, Ugt1a7, and Ugt2b1 were decreased in the HFD rats (Osabe et al. Citation2008). The alteration of Ugt1a1 and Ugt2b1 are not entirely consistent in animal models with NAFLD. Interestingly, the mRNA expression, protein expression, and activity of Ugt1a1, Ugt1a6, Ugt1a9, and Ugt2b7 in obesity, T2DM, or dyslipidemia showed down-regulated changes (Li et al. Citation2018; Zhang et al. Citation2019; Xu et al. Citation2022).

GSTs belong to a family of detoxifying enzymes that protect cellular macromolecules from reactive electrophilic reagents and contribute to the removal of toxic compounds from the cell. The GSTs genes expressed in the liver are GSTA1, GSTA2, GSTA4, GSTM1, and GSTM2 (Desmots et al. Citation2002; Kaur et al. Citation2020). GSTA1 favors the formation of the R-GSH conjugate of both prostaglandin A2 and prostaglandin J2. GSTA2 only showed some minor formation of the R-GSH conjugate of prostaglandin J2 (Bogaards et al. Citation1997). GSTA4 possesses high catalytic efficiency toward 4-hydroxynonenal (4-HNE), a cytotoxic end product of lipid peroxidation (Balogh et al. Citation2010). GSTM1 is involved in the biosynthesis of 14, 15-hepoxilins (Brunnström et al. Citation2011). GSTM2 has been shown to protect astrocytes from aminochrome-induced toxicity (Huenchuguala et al. Citation2016). In human and mouse models, the hepatic mRNA expression of GSTA1 was significantly increased (Lee et al. Citation2016; Lee, Park, et al. Citation2020; Xu et al. Citation2021). The mRNA expression of GSTA4 was up-regulated in mice with a high-fat diet (Sharma et al. Citation2018). It was found that GSTM2 mRNA and protein expression markedly decreased in NASH patients. Consistent with the clinical results, the mRNA and protein expression of Gstm2 were also significantly down-regulated in mice after a 24-week HFD, a 16-week HFHCD, or a 4-week MCDD (Lan et al. Citation2022). Moreover, the variation trend of GSTM2 in metabolic syndrome was consistent with that in NAFLD (Carreira et al. Citation2018).

SULTs are cytosolic enzymes that catalyze the sulfonation of endogenous and exogenous compounds by adding a sulfonate fraction to the compound to increase its water solubility and reduce its biological activity. In humans, SULTs are classified into five gene families, SULT1, SULT2, SULT3, SULT4, and SULT6 families (Kurogi et al. Citation2021). SULT1A1 is the main enzyme of SULTs in the liver, followed by SULT2A1, SULT1B1, and SULT1E1 (Riches et al. Citation2009). SULT1A1 sulfonates pro-carcinogens such as hydroxymethyl polycyclic aromatic hydrocarbons (Hempel et al. Citation2007). SULT2A1 is the major hydroxysteroid (alcohol) sulfotransferase, and it catalyzes the 3′-phosphoadenosine-5′-phosphosulfate-dependent sulfation of various endogenous hydroxysteroids as well as many xenobiotics that contain alcohol and phenol functional groups (Gulcan and Duffel Citation2011). SULT1B1 is involved in the sulfation of dotinurad (Omura et al. Citation2021). SULT1E1 plays an important role in estrogen homeostasis by sulfonating and deactivating estrogens (Silva Barbosa et al. Citation2020). In male C57BL/6 mice fed with the HFD, the mRNA expression of Sult1a1 was higher than those with a low-fat diet at 8 and 18 weeks (Wang et al. Citation2020). The study showed that the HFD decreased the Sult1b1 and Sult1e1 mRNA expression in mice (Wang, Tao, et al. Citation2016). However, the protein levels of Sult1e1 were up-regulated in the NAFLD rat model (Cong et al. Citation2021). The species, breeds, and genetic background of the animal models may be the factors that influence gene expression. The mRNA expression of SULT1A1 in rats with T2DM or dyslipidemia was down-regulated (Li et al. Citation2018; Xu et al. Citation2022). And Fashe et al. found the mRNA expression of SULT1E1 in patients with T2DM was up-regulated (Fashe et al. Citation2021). The above studies have reported that the metabolic comorbidities also regulated the expression of phase I/II DMEs and that some of the gene regulation trends are consistent with NAFLD.

5. The alteration of pharmacokinetic parameters in NAFLD

There is a new and more and more frequently applied tendency to use natural products in curing/alleviating NAFLD, such as spirulina, oleuropein, garlic, berberine, resveratrol, curcumin, ginseng, glycyrrhizin, coffee, cocoa powder, epigallocatechin-3-gallate, and bromelain. And many promising drug candidates are present in the current development pipeline that are of natural origin (Tarantino et al. Citation2021). Meanwhile, Chachay et al. speculated that resveratrol kinetics may be altered in NAFLD with obesity, resulting in reduced parent concentration (Chachay et al. Citation2014), which has prompted a movement to consider whether DMEs in NAFLD also affect pharmacokinetic parameters. Although few clinical studies have reported the impact of NAFLD on commonly prescribed drugs and natural products, they strongly emphasize the potential of NAFLD to cause variable drug responses and adverse drug reactions through altering pharmacokinetic parameters (Cobbina and Akhlaghi Citation2017). A limited number of pharmacokinetic studies are currently available on NAFLD. shows the substrates and pharmacokinetic parameters of the major DMEs whose expression was altered under the pathological conditions of NAFLD.

Table 4. Pharmacokinetic changes of drugs in NAFLD.

Clozapine, the substrate of CYP1A2, is an atypical antipsychotic. It is mainly used for the treatment of bipolar disorder and schizophrenia (Nucifora et al. Citation2017; Delgado et al. Citation2020). The main metabolite of clozapine is norclozapine. Norclozapine is a pharmacologically active compound and has more metabolic side effects than clozapine. In orotic acid diet (OAD) rats, consistent with previous reports, the hepatic protein expression of CYP1A2 was down-regulated, resulting in a significant increase in the area under curve (AUC) and AUCnorclozapine/AUCclozapine ratios of norclozapine. In addition, higher steady-state brain concentrations of clozapine and norclozapine were observed in OAD rats. These results suggested that caution may be warranted in the use of clozapine in patients with preexisting or drug-induced NAFLD (Li et al. Citation2021).

Caffeine is a naturally occurring stimulant found in coffee, tea, and chocolate. And it is used as an additive in other beverages and adjuvant analgesic in some pain medications (Mandel Citation2002; Sawynok Citation2011). Systemic caffeine clearance is considered the gold standard for phenotyping CYP1A2 in epidemiological studies. And the measurement of a unique salivary caffeine concentration has been recommended for verifying the presence and eventually the type of compensated liver cirrhosis. The results demonstrated that the salivary pharmacokinetics of caffeine in cirrhotic patients is very prolonged with respect to the findings of a previous study carried out in healthy subjects in which the mean serum half-life of caffeine was found to be very short. (Tarantino et al. Citation2006). Omeprazole, a proton pump inhibitor, is used to treat patients with erosive esophagitis who have gastroesophageal reflux disease (Richter et al. Citation2001). In ob/ob and ob/ob MCDD mice, the hepatic mRNA expression and protein levels of Cyp1a2, and Cyp2c29 (homologues human CYP2C19) were significantly decreased. Furthermore, the pharmacokinetics of caffeine and omeprazole, the probe drugs of Cyp1a2 and Cyp2c29, were significantly changed. The maximum concentration (Cmax) and the AUC of caffeine were significantly increased, while the AUCmetabolite/AUCdrug ratio of caffeine and omeprazole was significantly decreased in ob/ob mice, and ob/ob MCDD mice (Li, Clarke, et al. Citation2017). Consistently, in NASH adults and adolescents, the activity of CYP2C19 was a significant decrease. And the metabolic AUC ratio of omeprazole was decreased by 71% in NASH adolescents (Li, Canet, et al. Citation2017). These results revealed that plasma concentrations of caffeine and omeprazole are higher in patients with NAFLD at the same dose. It is unclear whether higher plasma concentrations of caffeine and omeprazole in patients with NAFLD at the same dose affect the adverse effects and clinical efficacy. And more objective and rigorous studies are required to determine whether the dosage of these two drugs can be adjusted in the NAFLD population.

Chlorzoxazone is a centrally-acting agent for painful musculoskeletal conditions (Nielsen et al. Citation2016), and it is a selective probe of CYP2E1 activity in vivo. It was found that the metabolic clearance of chlorzoxazone was markedly elevated in obese subjects with NASH than those without NASH (Emery et al. Citation2003). Midazolam is a short-acting benzodiazepine that is widely used as a premedicant prior to surgery, for induction of anesthesia, and for conscious sedation (Rodolà Citation2006). The CYP3A4 protein expression levels were significantly lower in patients with NAFLD and NASH. Compared with control subjects, midazolam concentrations were significantly higher in human subjects with NASH (Woolsey et al. Citation2015; Jamwal et al. Citation2018). In the HFD-fed mice model, both mRNA and protein expression levels of Cyp3a11/CYP3A4 tended to decrease. What’s more, HFD mice had significantly prolonged sleeping times after midazolam (Cyp3a11/CYP3A4 substrate) (Ghose et al. Citation2011). The results led us to wonder whether current drug dosage recommendations may need to be reevaluated in the NAFLD population to ensure the premier clinical outcomes of these drug therapies.

6. Future perspectives and conclusion

In conclusion, DMEs (phase I and II DMEs) play an important role in the metabolism of drugs. The altered expression of DMEs has been found in rodent and human samples with NAFLD and NAFLD-related metabolic comorbidities. The data of enzymatic activity, mRNA expression, and protein expression of the CYP isoforms are not in complete concordance with each other, which may be indicative of the heterogeneity in the regulation mechanisms known for each isoform. In addition, the species, breeds, and genetic background of the animal models may be the factors that influence gene expression. We summarized the pharmacokinetic changes of clozapine (CYP1A2 substrate), caffeine (CYP1A2 substrate), omeprazole (Cyp2c29/CYP2C19 substrate), chlorzoxazone (CYP2E1 substrate), midazolam (Cyp3a11/CYP3A4 substrate) in NAFLD. These results led us to wonder whether current drug dosage recommendations may need to be reevaluated in the NAFLD population to avoid serious adverse effects in patients with NAFLD. However, most studies about the alteration of pharmacokinetic parameters have been conducted in rodents. Therefore, the clinical applicability of the pharmacokinetic data is limited. More objective and rigorous studies are required to confirm these pharmacokinetic changes in patients with NAFLD and determine whether these pharmacokinetic changes affect drug dosage recommendations in the NAFLD population. At present, we have also summarized the substrates of the DMEs aforementioned to provide future research direction in . Thus, future investigations should focus on the effect and alteration of DMEs and pharmacokinetic parameters in this special patient population with NAFLD.

Table 5. List of well-known substrates for phase I and II DMEs.

Author contributions

Yan Zhu: Conceptualization, Writing- Reviewing and Editing. Li Chen: Investigation, Writing—Original Draft preparation. Yan Zhu and Li Chen had the same contribution to this work. Yuqi He: Supervision. Lin Qin: Visualization. Daopeng Tan: Visualization. Zhaojun Bai and Yu Song: Methodology. Yuhe Wang: Supervision and Project administration. All authors contributed to the article and approved the submitted version.

Disclosure statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Additional information

Funding

This work was supported by the [Science and Technology Program of Guizhou Province] under Grant [number QKHZC [2019]2829]; [Health Commission of Guizhou Province] under Grant [number gzwkj2022-220]; [Science and Technology Program of Guizhou Province] under Grant [number QKHZC [2022]-293]; [Science and Technology Program of Guizhou Province] under Grant [number QKHZC S[2020]2319].

References

  • Ahlström MM, Ridderström M, Zamora I, Luthman K. 2007. CYP2C9 structure-metabolism relationships: optimizing the metabolic stability of COX-2 inhibitors. J Med Chem. 50(18):4444–4452.
  • Almazroo OA, Miah MK, Venkataramanan R. 2017. Drug metabolism in the liver. Clin Liver Dis. 21(1):1–20.
  • Baiges-Gaya G, Fernández-Arroyo S, Luciano-Mateo F, Cabré N, Rodríguez-Tomàs E, Hernández-Aguilera A, Castañé H, Romeu M, Nogués M-R, Camps J, et al. 2021. Hepatic metabolic adaptation and adipose tissue expansion are altered in mice with steatohepatitis induced by high-fat high sucrose diet. J Nutr Biochem. 89:108559.
  • Balogh LM, Le Trong I, Kripps KA, Shireman LM, Stenkamp RE, Zhang W, Mannervik B, Atkins WM. 2010. Substrate specificity combined with stereopromiscuity in glutathione transferase A4-4-dependent metabolism of 4-hydroxynonenal. Biochemistry. 49(7):1541–1548.
  • Bock KW, Köhle C. 2005. UDP-glucuronosyltransferase 1A6: structural, functional, and regulatory aspects. Methods Enzymol. 400:57–75.
  • Bogaards JJ, Venekamp JC, van Bladeren PJ. 1997. Stereoselective conjugation of prostaglandin A2 and prostaglandin J2 with glutathione, catalyzed by the human glutathione S-transferases A1-1, A2-2, M1a-1a, and P1-1. Chem Res Toxicol. 10(3):310–317.
  • Borrie AE, Rose RV, Choi Y-H, Perera FE, Read N, Sexton T, Lock M, Vandenberg TA, Hahn K, Dinniwell R, et al. 2018. Letrozole concentration is associated with CYP2A6 variation but not with arthralgia in patients with breast cancer. Breast Cancer Res Treat. 172(2):371–379.
  • Brunnström A, Hamberg M, Griffiths WJ, Mannervik B, Claesson H-E. 2011. Biosynthesis of 14,15-hepoxilins in human l1236 Hodgkin lymphoma cells and eosinophils. Lipids. 46(1):69–79.
  • Buechler C, Weiss TS. 2011. Does hepatic steatosis affect drug metabolizing enzymes in the liver? Curr Drug Metab. 12(1):24–34.
  • Capi M, Curto M, Lionetto L, de Andrés F, Gentile G, Negro A, Martelletti P. 2016. Eletriptan in the management of acute migraine: an update on the evidence for efficacy, safety, and consistent response. Ther Adv Neurol Disord. 9(5):414–423.
  • Carreira MC, Izquierdo AG, Amil M, Casanueva FF, Crujeiras AB. 2018. Oxidative stress induced by excess of adiposity is related to a downregulation of hepatic SIRT6 expression in obese individuals. Oxid Med Cell Longev. 2018:6256052.
  • Chachay VS, Macdonald GA, Martin JH, Whitehead JP, O'Moore-Sullivan TM, Lee P, Franklin M, Klein K, Taylor PJ, Ferguson M, et al. 2014. Resveratrol does not benefit patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 12(12):2092–2103.e6.
  • Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, Charlton M, Sanyal AJ. 2012. The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Gastroenterological Association, American Association for the Study of Liver Diseases, and American College of Gastroenterology. Gastroenterology. 142(7):1592–1609.
  • Chang W-C, Jia H, Aw W, Saito K, Hasegawa S, Kato H. 2014. Beneficial effects of soluble dietary Jerusalem artichoke (Helianthus tuberosus) in the prevention of the onset of type 2 diabetes and non-alcoholic fatty liver disease in high-fructose diet-fed rats. Br J Nutr. 112(5):709–717.
  • Chiba T, Noji K, Shinozaki S, Suzuki S, Umegaki K, Shimokado K. 2016. Diet-induced non-alcoholic fatty liver disease affects expression of major cytochrome P450 genes in a mouse model. J Pharm Pharmacol. 68(12):1567–1576.
  • Cho Y-E, Kim D-K, Seo W, Gao B, Yoo S-H, Song B-J. 2021. Fructose promotes leaky gut, endotoxemia, and liver fibrosis through ethanol-inducible cytochrome P450-2E1-mediated oxidative and nitrative stress. Hepatology. 73(6):2180–2195.
  • Cobbina E, Akhlaghi F. 2017. Non-alcoholic fatty liver disease (NAFLD) - pathogenesis, classification, and effect on drug metabolizing enzymes and transporters. Drug Metab Rev. 49(2):197–211.
  • Cong S, Li Z, Yu L, Liu Y, Hu Y, Bi Y, Cheng M. 2021. Integrative proteomic and lipidomic analysis of Kaili Sour Soup-mediated attenuation of high-fat diet-induced nonalcoholic fatty liver disease in a rat model. Nutr Metab. 18(1):26.
  • Connor F, Rayner TF, Aitken SJ, Feig C, Lukk M, Santoyo-Lopez J, Odom DT. 2018. Mutational landscape of a chemically-induced mouse model of liver cancer. J Hepatol. 69(4):840–850.
  • Cook I, Wang T, Leyh TS. 2019. Isoform-specific therapeutic control of sulfonation in humans. Biochem Pharmacol. 159:25–31.
  • Court MH, Hazarika S, Krishnaswamy S, Finel M, Williams JA. 2008. Novel polymorphic human UDP-glucuronosyltransferase 2A3: cloning, functional characterization of enzyme variants, comparative tissue expression, and gene induction. Mol Pharmacol. 74(3):744–754.
  • Danielson PB. 2002. The cytochrome P450 superfamily: biochemistry, evolution and drug metabolism in humans. Curr Drug Metab. 3(6):561–597.
  • Delgado A, Velosa J, Zhang J, Dursun SM, Kapczinski F, de Azevedo Cardoso T. 2020. Clozapine in bipolar disorder: a systematic review and meta-analysis. J Psychiatr Res. 125:21–27.
  • Desmots F, Rissel M, Pigeon C, Loyer P, Loréal O, Guillouzo A. 2002. Differential effects of iron overload on GST isoform expression in mouse liver and kidney and correlation between GSTA4 induction and overproduction of free radicles. Free Radic Biol Med. 32(1):93–101.
  • Duthaler U, Bachmann F, Suenderhauf C, Grandinetti T, Pfefferkorn F, Haschke M, Hruz P, Bouitbir J, Krähenbühl S. 2022. Liver cirrhosis affects the pharmacokinetics of the six substrates of the basel phenotyping cocktail differently. Clin Pharmacokinet. 61(7):1039–1055.
  • El Daibani AA, Alherz FA, Abunnaja MS, Bairam AF, Rasool MI, Kurogi K, Liu M-C. 2021. Impact of human SULT1E1 polymorphisms on the sulfation of 17β-Estradiol, 4-hydroxytamoxifen, and diethylstilbestrol by SULT1E1 allozymes. Eur J Drug Metab Pharmacokinet. 46(1):105–118.
  • Emery MG, Fisher JM, Chien JY, Kharasch ED, Dellinger EP, Kowdley KV, Thummel KE. 2003. CYP2E1 activity before and after weight loss in morbidly obese subjects with nonalcoholic fatty liver disease. Hepatology. 38(2):428–435.
  • Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, Zelber-Sagi S, Wai-Sun Wong V, Dufour J-F, Schattenberg JM, et al. 2020. A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J Hepatol. 73(1):202–209.
  • Faber MS, Jetter A, Fuhr U. 2005. Assessment of CYP1A2 activity in clinical practice: why, how, and when? Basic Clin Pharmacol Toxicol. 97(3):125–134.
  • Fashe M, Yi M, Sueyoshi T, Negishi M. 2021. Sex-specific expression mechanism of hepatic estrogen inactivating enzyme and transporters in diabetic women. Biochem Pharmacol. 190:114662.
  • Fisher CD, Lickteig AJ, Augustine LM, Ranger-Moore J, Jackson JP, Ferguson SS, Cherrington NJ. 2009. Hepatic cytochrome P450 enzyme alterations in humans with progressive stages of nonalcoholic fatty liver disease. Drug Metab Dispos. 37(10):2087–2094.
  • Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. 2018. Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 24(7):908–922.
  • Fuhr LM, Marok FZ, Mees M, Mahfoud F, Selzer D, Lehr T. 2022. A physiologically based pharmacokinetic and pharmacodynamic model of the CYP3A4 substrate felodipine for drug-drug interaction modeling. Pharmaceutics. 14(7):1474.
  • Gagné J-F, Montminy V, Belanger P, Journault K, Gaucher G, Guillemette C. 2002. Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Mol Pharmacol. 62(3):608–617.
  • Gao Y, Zhang W, Zeng L-Q, Bai H, Li J, Zhou J, Zhou G-Y, Fang C-W, Wang F, Qin X-J. 2020. Exercise and dietary intervention ameliorate high-fat diet-induced NAFLD and liver aging by inducing lipophagy. Redox Biol. 36:101635.
  • Ghose R, Omoluabi O, Gandhi A, Shah P, Strohacker K, Carpenter KC, McFarlin B, Guo T. 2011. Role of high-fat diet in regulation of gene expression of drug metabolizing enzymes and transporters. Life Sci. 89(1-2):57–64.
  • Goetz MP, Sangkuhl K, Guchelaar H-J, Schwab M, Province M, Whirl-Carrillo M, Symmans WF, McLeod HL, Ratain MJ, Zembutsu H, et al. 2018. Clinical pharmacogenetics implementation consortium (CPIC) guideline for CYP2D6 and tamoxifen therapy. Clin Pharmacol Ther. 103(5):770–777.
  • Gofton C, Upendran Y, Zheng M-H, George J. 2022. MAFLD: what is different from NAFLD? Clin Mol Hepatol.29(Suppl):S17–S31.
  • Gradinaru J, Romand S, Geiser L, Carrupt P-A, Spaggiari D, Rudaz S. 2015. Inhibition screening method of microsomal UGTs using the cocktail approach. Eur J Pharm Sci. 71:35–45.
  • Griesel R, Maartens G, Chirehwa M, Sokhela S, Akpomiemie G, Moorhouse M, Venter F, Sinxadi P. 2021. CYP2B6 genotype and weight gain differences between dolutegravir and efavirenz. Clin Infect Dis. 73(11):e3902–e3909.
  • Gulcan HO, Duffel MW. 2011. Substrate inhibition in human hydroxysteroid sulfotransferase SULT2A1: studies on the formation of catalytically non-productive enzyme complexes. Arch Biochem Biophys. 507(2):232–240.
  • Guo Y, Li F, Ma X, Cheng X, Zhou H, Klaassen CD. 2011. CYP2D plays a major role in berberine metabolism in liver of mice and humans. Xenobiotica. 41(11):996–1005.
  • Hao H, Lai L, Zheng C, Wang Q, Yu G, Zhou X, Wu L, Gong P, Wang G. 2010. Microsomal cytochrome p450-mediated metabolism of protopanaxatriol ginsenosides: metabolite profile, reaction phenotyping, and structure-metabolism relationship. Drug Metab Dispos. 38(10):1731–1739.
  • Hardwick RN, Ferreira DW, More VR, Lake AD, Lu Z, Manautou JE, Slitt AL, Cherrington NJ. 2013. Altered UDP-glucuronosyltransferase and sulfotransferase expression and function during progressive stages of human nonalcoholic fatty liver disease. Drug Metab Dispos. 41(3):554–561.
  • Hempel N, Gamage N, Martin JL, McManus ME. 2007. Human cytosolic sulfotransferase SULT1A1. Int J Biochem Cell Biol. 39(4):685–689.
  • Henry L, Paik J, Younossi ZM. 2022. Review article: the epidemiologic burden of non-alcoholic fatty liver disease across the world. Aliment Pharmacol Ther. 56(6):942–956.
  • Hoehle SI, Pfeiffer E, Metzler M. 2007. Glucuronidation of curcuminoids by human microsomal and recombinant UDP-glucuronosyltransferases. Mol Nutr Food Res. 51(8):932–938.
  • Hong CH, Ko MS, Kim JH, Cho H, Lee C-H, Yoon JE, Yun J-Y, Baek I-J, Jang JE, Lee SE, et al. 2022. Sphingosine 1-phosphate receptor 4 promotes nonalcoholic steatohepatitis by activating NLRP3 inflammasome. Cell Mol Gastroenterol Hepatol. 13(3):925–947.
  • Hua YQ, Zeng Y, Xu J, Xu XL. 2021. Naringenin alleviates nonalcoholic steatohepatitis in middle-aged Apoemice: role of SIRT1. Phytomedicine. 81:153412.
  • Huang S, Wu Y, Zhao Z, Wu B, Sun K, Wang H, Qin L, Bai F, Leng Y, Tang W. 2021. A new mechanism of obeticholic acid on NASH treatment by inhibiting NLRP3 inflammasome activation in macrophage. Metabolism. 120:154797.
  • Huang X, Li C, Li C, Li Z, Li X, Liao J, Rao T, Chen L, Gao L, Ouyang D. 2021. CYP2C19 genotyping may provide a better treatment strategy when administering escitalopram in Chinese population. Front Pharmacol. 12:730461.
  • Huenchuguala S, Muñoz P, Graumann R, Paris I, Segura-Aguilar J. 2016. DT-diaphorase protects astrocytes from aminochrome-induced toxicity. Neurotoxicology. 55:10–12.
  • Jamwal R, de la Monte SM, Ogasawara K, Adusumalli S, Barlock BB, Akhlaghi F. 2018. Nonalcoholic fatty liver disease and diabetes are associated with decreased CYP3A4 protein expression and activity in human liver. Mol Pharm. 15(7):2621–2632.
  • Jaworski M, Buchmann A, Bauer P, Riess O, Schwarz M. 2005. B-raf and Ha-ras mutations in chemically induced mouse liver tumors. Oncogene. 24(7):1290–1295.
  • Jian T, Yu C, Ding X, Chen J, Li J, Zuo Y, Ren B, Lv H, Li W. 2019. Hepatoprotective effect of seed coat of Euryale ferox extract in non-alcoholic fatty liver disease induced by high-fat diet in mice by increasing IRs-1 and inhibiting CYP2E1. J Oleo Sci. 68(6):581–589.
  • Kasichayanula S, Liu X, Griffen SC, Lacreta FP, Boulton DW. 2013. Effects of rifampin and mefenamic acid on the pharmacokinetics and pharmacodynamics of dapagliflozin. Diabetes Obes Metab. 15(3):280–283.
  • Kaur G, Gupta SK, Singh P, Ali V, Kumar V, Verma M. 2020. Drug-metabolizing enzymes: role in drug resistance in cancer. Clin Transl Oncol. 22(10):1667–1680.
  • Kim TH, Choi D, Kim JY, Lee JH, Koo S-H. 2017. Fast food diet-induced non-alcoholic fatty liver disease exerts early protective effect against acetaminophen intoxication in mice. BMC Gastroenterol. 17(1):124.
  • Koch KM, Corrigan BW, Manzo J, James CD, Scott RJ, Stead AG, Kersey KE. 2004. Alosetron repeat dose pharmacokinetics, effects on enzyme activities, and influence of demographic factors. Aliment Pharmacol Ther. 20(2):223–230.
  • Konopnicki CM, Dickmann LJ, Tracy JM, Tukey RH, Wienkers LC, Foti RS. 2013. Evaluation of UGT protein interactions in human hepatocytes: effect of siRNA down regulation of UGT1A9 and UGT2B7 on propofol glucuronidation in human hepatocytes. Arch Biochem Biophys. 535(2):143–149.
  • Kumar A, Sundaram K, Mu J, Dryden GW, Sriwastva MK, Lei C, Zhang L, Qiu X, Xu F, Yan J, et al. 2021. High-fat diet-induced upregulation of exosomal phosphatidylcholine contributes to insulin resistance. Nat Commun. 12(1):213.
  • Kurogi K, Rasool MI, Alherz FA, El Daibani AA, Bairam AF, Abunnaja MS, Yasuda S, Wilson LJ, Hui Y, Liu M-C. 2021. SULT genetic polymorphisms: physiological, pharmacological and clinical implications. Expert Opin Drug Metab Toxicol. 17(7):767–784.
  • Kvitne KE, Åsberg A, Johnson LK, Wegler C, Hertel JK, Artursson P, Karlsson C, Andersson S, Sandbu R, Skovlund E, et al. 2022. Impact of type 2 diabetes on in vivo activities and protein expressions of cytochrome P450 in patients with obesity. Clin Transl Sci. 15(11):2685–2696.
  • Lad A, Hunyadi J, Connolly J, Breidenbach JD, Khalaf FK, Dube P, Zhang S, Kleinhenz AL, Baliu-Rodriguez D, Isailovic D, et al. 2022. Antioxidant therapy significantly attenuates hepatotoxicity following low dose exposure to microcystin-LR in a murine model of diet-induced non-alcoholic fatty liver disease. Antioxidants. 11(8):1625.
  • Lan T, Hu Y, Hu F, Li H, Chen Y, Zhang J, Yu Y, Jiang S, Weng Q, Tian S, et al. 2022. Hepatocyte glutathione S-transferase mu 2 prevents non-alcoholic steatohepatitis by suppressing ASK1 signaling. J Hepatol. 76(2):407–419.
  • Lee DH, Han DH, Nam KT, Park JS, Kim SH, Lee M, Kim G, Min BS, Cha B-S, Lee YS, et al. 2016. Ezetimibe, an NPC1L1 inhibitor, is a potent Nrf2 activator that protects mice from diet-induced nonalcoholic steatohepatitis. Free Radic Biol Med. 99:520–532.
  • Lee DH, Park JS, Lee YS, Han J, Lee D-K, Kwon SW, Han DH, Lee Y-H, Bae SH. 2020. SQSTM1/p62 activates NFE2L2/NRF2 via ULK1-mediated autophagic KEAP1 degradation and protects mouse liver from lipotoxicity. Autophagy. 16(11):1949–1973.
  • Lee ES, Kwon M-H, Kim HM, Woo HB, Ahn CM, Chung CH. 2020. Curcumin analog CUR5-8 ameliorates nonalcoholic fatty liver disease in mice with high-fat diet-induced obesity. Metabolism. 103:154015.
  • Lee J-T, Pao L-H, Hsiong C-H, Huang P-W, Shih T-Y, Yoa-Pu Hu O. 2013. Validated liquid chromatography-tandem mass spectrometry method for determination of totally nine probe metabolites of cytochrome P450 enzymes and UDP-glucuronosyltransferases. Talanta. 106:220–228.
  • Lépine J, Bernard O, Plante M, Têtu B, Pelletier G, Labrie F, Bélanger A, Guillemette C. 2004. Specificity and regioselectivity of the conjugation of estradiol, estrone, and their catecholestrogen and methoxyestrogen metabolites by human uridine diphospho-glucuronosyltransferases expressed in endometrium. J Clin Endocrinol Metab. 89(10):5222–5232.
  • Lévesque E, Girard H, Journault K, Lépine J, Guillemette C. 2007. Regulation of the UGT1A1 bilirubin-conjugating pathway: role of a new splicing event at the UGT1A locus. Hepatology. 45(1):128–138.
  • Li H, Canet MJ, Clarke JD, Billheimer D, Xanthakos SA, Lavine JE, Erickson RP, Cherrington NJ. 2017. Pediatric cytochrome P450 activity alterations in nonalcoholic steatohepatitis. Drug Metab Dispos. 45(12):1317–1325.
  • Li H, Clarke JD, Dzierlenga AL, Bear J, Goedken MJ, Cherrington NJ. 2017. In vivo cytochrome P450 activity alterations in diabetic nonalcoholic steatohepatitis mice. J Biochem Mol Toxicol. 31(2):e21840.
  • Li P, Robertson TA, Thorling CA, Zhang Q, Fletcher LM, Crawford DHG, Roberts MS. 2011. Hepatic pharmacokinetics of cationic drugs in a high-fat emulsion-induced rat model of nonalcoholic steatohepatitis. Drug Metab Dispos. 39(4):571–579.
  • Li R, Guo G, Cao Y, Wang Y, Liu F, Zhang X. 2013. Expression of cytochrome P450 2A5 in a C57BL/6J mouse model of nonalcoholic fatty liver disease. Pharmacology. 92(1-2):26–31.
  • Li X, Li S, Chen M, Wang J, Xie B, Sun Z. 2018. (-)-Epigallocatechin-3-gallate (EGCG) inhibits starch digestion and improves glucose homeostasis through direct or indirect activation of PXR/CAR-mediated phase II metabolism in diabetic mice. Food Funct. 9(9):4651–4663.
  • Li Z, Lee SH, Jeong HJ, Kang HE. 2021. Pharmacokinetic changes of clozapine and norclozapine in a rat model of non-alcoholic fatty liver disease induced by orotic acid. Xenobiotica. 51(3):324–334.
  • Lin Q-M, Li Y-H, Lu X-R, Wang R, Pang N-H, Xu R-A, Cai J-P, Hu G-X. 2019. Characterization of genetic variation in CYP3A4 on the metabolism of cabozantinib in vitro. Chem Res Toxicol. 32(8):1583–1590.
  • Lin S, Huang J, Wang M, Kumar R, Liu Y, Liu S, Wu Y, Wang X, Zhu Y. 2020. Comparison of MAFLD and NAFLD diagnostic criteria in real world. Liver Int. 40(9):2082–2089.
  • Liu W, Shang J, Deng Y, Han X, Chen Y, Wang S, Yang R, Dong F, Shang H. 2022. Network pharmacology analysis on mechanism of Jian Pi Qing Gan Yin decoction ameliorating high fat diet-induced non-alcoholic fatty liver disease and validated in vivo. J Ethnopharmacol. 295:115382.
  • Liu Y, Deng J, Fan D. 2019. Ginsenoside Rk3 ameliorates high-fat-diet/streptozocin induced type 2 diabetes mellitus in mice via the AMPK/Akt signaling pathway. Food Funct. 10(5):2538–2551.
  • Liu Y, Xu W, Zhai T, You J, Chen Y. 2019. Silibinin ameliorates hepatic lipid accumulation and oxidative stress in mice with non-alcoholic steatohepatitis by regulating CFLAR-JNK pathway. Acta Pharm Sin B. 9(4):745–757.
  • Locuson CW, Williams P, Adcock JM, Daniels JS. 2016. Evaluation of tizanidine as a marker of canine CYP1A2 activity. J Vet Pharmacol Ther. 39(2):122–130.
  • Lu Y, Zhu J, Chen X, Li N, Fu F, He J, Wang G, Zhang L, Zheng Y, Qiu Z, et al. 2009. Identification of human UDP-glucuronosyltransferase isoforms responsible for the glucuronidation of glycyrrhetinic acid. Drug Metab Pharmacokinet. 24(6):523–528.
  • Maksymchuk O, Shysh A, Stroy D. 2022. Treatment with omega-3 PUFAs does not increase the risk of CYP2E1-dependent oxidative stress and diabetic liver pathology. Front Endocrinol. 13:1004564.
  • Mandel HG. 2002. Update on caffeine consumption, disposition and action. Food Chem Toxicol. 40(9):1231–1234.
  • Marok FZ, Fuhr LM, Hanke N, Selzer D, Lehr T. 2021. Physiologically based pharmacokinetic modeling of bupropion and its metabolites in a CYP2B6 drug-drug-gene interaction network. Pharmaceutics. 13(3):331.
  • Márquez-Quiroga LV, Arellanes-Robledo J, Vásquez-Garzón VR, Villa-Treviño S, Muriel P. 2022. Models of nonalcoholic steatohepatitis potentiated by chemical inducers leading to hepatocellular carcinoma. Biochem Pharmacol. 195:114845.
  • Matsumoto Y, Fujita S, Yamagishi A, Shirai T, Maeda Y, Suzuki T, Kobayashi K-I, Inoue J, Yamamoto Y. 2021. Brown rice inhibits development of nonalcoholic fatty liver disease in obese Zucker (fa/fa) rats by increasing lipid oxidation via activation of retinoic acid synthesis. J Nutr. 151(9):2705–2713.
  • Meech R, Hu DG, McKinnon RA, Mubarokah SN, Haines AZ, Nair PC, Rowland A, Mackenzie PI. 2019. The UDP-glycosyltransferase (UGT) superfamily: new members, new functions, and novel paradigms. Physiol Rev. 99(2):1153–1222.
  • Miura M, Uchida S, Tanaka S, Inui N, Kawakami J, Watanabe H, Namiki N. 2019. The prediction of the area under the curve and clearance of midazolam from single-point plasma concentration and urinary excretion in healthy volunteers. Biol Pharm Bull. 42(9):1590–1595.
  • Miyata M, Funaki A, Fukuhara C, Sumiya Y, Sugiura Y. 2020. Taurine attenuates hepatic steatosis in a genetic model of fatty liver disease. J Toxicol Sci. 45(2):87–94.
  • Miyata M, Matsushita K, Shindo R, Shimokawa Y, Sugiura Y, Yamashita M. 2020. Selenoneine ameliorates hepatocellular injury and hepatic steatosis in a mouse model of NAFLD. Nutrients. 12(6):1898.
  • Nakanishi K, Kaji K, Kitade M, Kubo T, Furukawa M, Saikawa S, Shimozato N, Sato S, Seki K, Kawaratani H, et al. 2019. Exogenous administration of low-dose lipopolysaccharide potentiates liver fibrosis in a choline-deficient l-amino-acid-defined diet-induced murine steatohepatitis model. Int J Mol Sci. 20(11). DOI:10.3390/ijms20112724
  • Negi CK, Babica P, Bajard L, Bienertova-Vasku J, Tarantino G. 2022. Insights into the molecular targets and emerging pharmacotherapeutic interventions for nonalcoholic fatty liver disease. Metabolism. 126:154925.
  • Nielsen RV, Fomsgaard JS, Siegel H, Martusevicius R, Mathiesen O, Dahl JB. 2016. The effect of chlorzoxazone on acute pain after spine surgery. A randomized, blinded trial. Acta Anaesthesiol Scand. 60(8):1152–1160.
  • Niu Z, Qiang T, Lin W, Li Y, Wang K, Wang D, Wang X. 2022. Evaluation of potential herb-drug interactions between Shengmai injection and losartan potassium in rat and in vitro. Front Pharmacol. 13:878526.
  • Nucifora FC, Mihaljevic M, Lee BJ, Sawa A. 2017. Clozapine as a model for antipsychotic development. Neurotherapeutics. 14(3):750–761.
  • Oh K-S, Park S-J, Shinde DD, Shin J-G, Kim D-H. 2012. High-sensitivity liquid chromatography-tandem mass spectrometry for the simultaneous determination of five drugs and their cytochrome P450-specific probe metabolites in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 895-896:56–64.
  • Omura K, Motoki K, Kobashi S, Miyata K, Yamano K, Iwanaga T. 2021. Identification of human UDP-glucuronosyltransferase and sulfotransferase as responsible for the metabolism of dotinurad, a novel selective urate reabsorption inhibitor. Drug Metab Dispos. 49(11):1016–1024.
  • Osabe M, Sugatani J, Fukuyama T, Ikushiro S-i, Ikari A, Miwa M. 2008. Expression of hepatic UDP-glucuronosyltransferase 1A1 and 1A6 correlated with increased expression of the nuclear constitutive androstane receptor and peroxisome proliferator-activated receptor alpha in male rats fed a high-fat and high-sucrose diet. Drug Metab Dispos. 36(2):294–302.
  • Oseini AM, Sanyal AJ. 2017. Therapies in non-alcoholic steatohepatitis (NASH). Liver Int. 37:97–103.
  • Pathania S, Bhatia R, Baldi A, Singh R, Rawal RK. 2018. Drug metabolizing enzymes and their inhibitors’ role in cancer resistance. Biomed Pharmacother. 105:53–65.
  • Piver B, Fer M, Vitrac X, Merillon J-M, Dreano Y, Berthou F, Lucas D. 2004. Involvement of cytochrome P450 1A2 in the biotransformation of trans-resveratrol in human liver microsomes. Biochem Pharmacol. 68(4):773–782.
  • Polyzos SA, Kang ES, Boutari C, Rhee E-J, Mantzoros CS. 2020. Current and emerging pharmacological options for the treatment of nonalcoholic steatohepatitis. Metabolism. 111S:154203.
  • Porras D, Nistal E, Martínez-Flórez S, Pisonero-Vaquero S, Olcoz JL, Jover R, González-Gallego J, García-Mediavilla MV, Sánchez-Campos S. 2017. Protective effect of quercetin on high-fat diet-induced non-alcoholic fatty liver disease in mice is mediated by modulating intestinal microbiota imbalance and related gut-liver axis activation. Free Radic Biol Med. 102:188–202.
  • Ranaweera SS, Natraj P, Rajan P, Dayarathne LA, Mihindukulasooriya SP, Dinh DTT, Jee Y, Han C-H. 2022. Anti-obesity effect of sulforaphane in broccoli leaf extract on 3T3-L1 adipocytes and ob/ob mice. J Nutr Biochem. 100:108885.
  • Riches Z, Stanley EL, Bloomer JC, Coughtrie MWH. 2009. Quantitative evaluation of the expression and activity of five major sulfotransferases (SULTs) in human tissues: the SULT “pie.” Drug Metab Dispos. 37(11):2255–2261.
  • Richter JE, Kahrilas PJ, Johanson J, Maton P, Breiter JR, Hwang C, Marino V, Hamelin B, Levine JG. 2001. Efficacy and safety of esomeprazole compared with omeprazole in GERD patients with erosive esophagitis: a randomized controlled trial. Am J Gastroenterology. 96(3):656–665.
  • Rodolà F. 2006. Midazolam as an anti-emetic. Eur Rev Med Pharmacol Sci. 10(3):121–126.
  • Sakurai Y, Kubota N, Yamauchi T, Kadowaki T. 2021. Role of insulin resistance in MAFLD. Int J Mol Sci. 22(8). DOI:10.3390/ijms22084156
  • Sawynok J. 2011. Methylxanthines and pain. Handb Exp Pharmacol. 200:311–329.
  • Sharma L, Gupta D, Abdullah ST. 2019. Thioacetamide potentiates high cholesterol and high fat diet induced steato-hepatitic changes in livers of C57BL/6J mice: a novel eight weeks model of fibrosing NASH. Toxicol Lett. 304:21–29.
  • Sharma RS, Harrison DJ, Kisielewski D, Cassidy DM, McNeilly AD, Gallagher JR, Walsh SV, Honda T, McCrimmon RJ, Dinkova-Kostova AT, et al. 2018. Experimental nonalcoholic steatohepatitis and liver fibrosis are ameliorated by pharmacologic activation of Nrf2 (NF-E2 p45-related factor 2). Cell Mol Gastroenterol Hepatol. 5(3):367–398.
  • Silva Barbosa AC, Zhou D, Xie Y, Choi Y-J, Tung H-C, Chen X, Xu M, Gibbs RB, Poloyac SM, Liu S, et al. 2020. Inhibition of estrogen sulfotransferase (/EST) ameliorates ischemic acute kidney injury in mice. J Am Soc Nephrol. 31(7):1496–1508.
  • Siu YA, Hao M-H, Dixit V, Lai WG. 2018. Celecoxib is a substrate of CYP2D6: impact on celecoxib metabolism in individuals with CYP2C9*3 variants. Drug Metab Pharmacokinet. 33(5):219–227.
  • Song Y, Li C, Liu G, Liu R, Chen Y, Li W, Cao Z, Zhao B, Lu C, Liu Y. 2021. Drug-metabolizing cytochrome P450 enzymes have multifarious influences on treatment outcomes. Clin Pharmacokinet. 60(5):585–601.
  • Stowers SJ, Wiseman RW, Ward JM, Miller EC, Miller JA, Anderson MW, Eva A. 1988. Detection of activated proto-oncogenes in N-nitrosodiethylamine-induced liver tumors: a comparison between B6C3F1 mice and Fischer 344 rats. Carcinogenesis. 9(2):271–276.
  • Suga T, Yamaguchi H, Ogura J, Shoji S, Maekawa M, Mano N. 2019. Altered bile acid composition and disposition in a mouse model of non-alcoholic steatohepatitis. Toxicol Appl Pharmacol. 379:114664.
  • Suzuki S, Nishijima C, Sato Y, Umegaki K, Murata M, Chiba T. 2020. Coleus forskohlii extract attenuated the beneficial effect of diet-treatment on NASH in mouse model. J Nutr Sci Vitaminol. 66(2):191–199.
  • Sylvester Darvin S, Toppo E, Esakkimuthu S, Ajeesh Krishna TP, Ceasar SA, Stalin A, Balakrishna K, Muniappan N, Pazhanivel N, Mahaprabhu R, et al. 2018. Hepatoprotective effect of bisbenzylisoquinoline alkaloid tiliamosine from Tiliacora racemosa in high-fat diet/diethylnitrosamine-induced non-alcoholic steatohepatitis. Biomed Pharmacother. 108:963–973.
  • Takahashi Y, Soejima Y, Fukusato T. 2012. Animal models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J Gastroenterol. 18(19):2300–2308.
  • Tarantino G, Balsano C, Santini SJ, Brienza G, Clemente I, Cosimini B, Sinatti G. 2021. It is high time physicians thought of natural products for alleviating NAFLD. Is there sufficient evidence to use them? Int J Mol Sci. 22(24). DOI:10.3390/ijms222413424
  • Tarantino G, Conca P, Capone D, Gentile A, Polichetti G, Basile V. 2006. Reliability of total overnight salivary caffeine assessment (TOSCA) for liver function evaluation in compensated cirrhotic patients. Eur J Clin Pharmacol. 62(8):605–612.
  • Tsuchida T, Lee YA, Fujiwara N, Ybanez M, Allen B, Martins S, Fiel MI, Goossens N, Chou H-I, Hoshida Y, et al. 2018. A simple diet- and chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J Hepatol. 69(2):385–395.
  • Tung Y-T, Huang C-Z, Lin J-H, Yen G-C. 2018. Effect of Phyllanthus emblica L. fruit on methionine and choline-deficiency diet-induced nonalcoholic steatohepatitis. J Food Drug Anal. 26(4):1245–1252.
  • van Oijen MGH, Huybers S, Peters WHM, Drenth JPH, Laheij RJF, Verheugt FWA, Jansen JBMJ. 2005. Polymorphisms in genes encoding acetylsalicylic acid metabolizing enzymes are unrelated to upper gastrointestinal health in cardiovascular patients on acetylsalicylic acid. Br J Clin Pharmacol. 60(6):623–628.
  • Verbeeck RK. 2008. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 64(12):1147–1161.
  • Vogel CFA, Van Winkle LS, Esser C, Haarmann-Stemmann T. 2020. The aryl hydrocarbon receptor as a target of environmental stressors - implications for pollution mediated stress and inflammatory responses. Redox Biol. 34:101530.
  • Wahlang B, Song M, Beier JI, Cameron Falkner K, Al-Eryani L, Clair HB, Prough RA, Osborne TS, Malarkey DE, States JC, et al. 2014. Evaluation of Aroclor 1260 exposure in a mouse model of diet-induced obesity and non-alcoholic fatty liver disease. Toxicol Appl Pharmacol. 279(3):380–390.
  • Wang C, Tao Q, Wang X, Wang X, Zhang X. 2016. Impact of high-fat diet on liver genes expression profiles in mice model of nonalcoholic fatty liver disease. Environ Toxicol Pharmacol. 45:52–62.
  • Wang J, Zhai T, Chen Y. 2018. Effects of honokiol on CYP450 activity and transporter mRNA expression in type 2 diabetic rats. Int J Mol Sci. 19(3). DOI:10.3390/ijms19030815
  • Wang K, Chen X, Ward SC, Liu Y, Ouedraogo Y, Xu C, Cederbaum AI, Lu Y. 2019. CYP2A6 is associated with obesity: studies in human samples and a high fat diet mouse model. Int J Obes. 43(3):475–486.
  • Wang P, Shao X, Bao Y, Zhu J, Chen L, Zhang L, Ma X, Zhong X-B. 2020. Impact of obese levels on the hepatic expression of nuclear receptors and drug-metabolizing enzymes in adult and offspring mice. Acta Pharm Sin B. 10(1):171–185.
  • Wang X-H, Cui X-X, Sun X-Q, Wang X-H, Li X-C, Qi Y, Li W, Han M-Y, Muhammad I, Zhang X-Y. 2017. High fat diet-induced hepatic 18-carbon fatty acids accumulation up-regulates CYP2A5/CYP2A6 via NF-E2-related factor 2. Front Pharmacol. 8:233.
  • Wang Y, You Y, Tian Y, Sun H, Li X, Wang X, Wang Y, Liu J. 2020. Pediococcus pentosaceus PP04 Ameliorates high-fat diet-induced hyperlipidemia by regulating lipid metabolism in C57BL/6N mice. J Agric Food Chem. 68(51):15154–15163.
  • Wang Y-C, Hsieh T-C, Chou C-L, Wu J-L, Fang T-C. 2016. Risks of adverse events following coprescription of statins and calcium channel blockers: a nationwide population-based study. Medicine. 95(2):e2487.
  • Wang Z, Wong T, Hashizume T, Dickmann LZ, Scian M, Koszewski NJ, Goff JP, Horst RL, Chaudhry AS, Schuetz EG, et al. 2014. Human UGT1A4 and UGT1A3 conjugate 25-hydroxyvitamin D3: metabolite structure, kinetics, inducibility, and interindividual variability. Endocrinology. 155(6):2052–2063.
  • Wegman P, Vainikka L, Stål O, Nordenskjöld B, Skoog L, Rutqvist L-E, Wingren S. 2005. Genotype of metabolic enzymes and the benefit of tamoxifen in postmenopausal breast cancer patients. Breast Cancer Res. 7(3):R284–R290.
  • Woolsey SJ, Mansell SE, Kim RB, Tirona RG, Beaton MD. 2015. CYP3A activity and expression in nonalcoholic fatty liver disease. Drug Metab Dispos. 43(10):1484–1490.
  • Wu J, Lou Y-G, Yang X-l, Wang R, Zhang R, Aa J-Y, Wang G-J, Xie Y. 2023. Silybin regulates P450s activity by attenuating endoplasmic reticulum stress in mouse nonalcoholic fatty liver disease. Acta Pharmacol Sin. 44(1):133–144.
  • Wu Z, Zhang Y, Gong X, Cheng G, Pu S, Cai S. 2020. The preventive effect of phenolic-rich extracts from Chinese sumac fruits against nonalcoholic fatty liver disease in rats induced by a high-fat diet. Food Funct. 11(1):799–812.
  • Xu J, Kulkarni SR, Li L, Slitt AL. 2012. UDP-glucuronosyltransferase expression in mouse liver is increased in obesity- and fasting-induced steatosis. Drug Metab Dispos. 40(2):259–266.
  • Xu J, Peng Y, Zeng Y, Hua Y-Q, Xu X-l. 2019. 2, 3, 4', 5-Tetrahydroxystilbene-2-0-β-d glycoside attenuates age- and diet-associated non-alcoholic steatohepatitis and atherosclerosis in LDL receptor knockout mice and its possible mechanisms. Int J Mol Sci. 20(7). DOI:10.3390/ijms20071617
  • Xu L, Yin L, Qi Y, Tan X, Gao M, Peng J. 2021. 3D disorganization and rearrangement of genome provide insights into pathogenesis of NAFLD by integrated Hi-C, Nanopore, and RNA sequencing. Acta Pharm Sin B. 11(10):3150–3164.
  • Xu Y, Lu J, Guo Y, Zhang Y, Liu J, Huang S, Zhang Y, Gao L, Wang X. 2022. Hypercholesterolemia reduces the expression and function of hepatic drug metabolizing enzymes and transporters in rats. Toxicol Lett. 364:1–11.
  • Yan C, Zhang Y, Zhang X, Aa J, Wang G, Xie Y. 2018. Curcumin regulates endogenous and exogenous metabolism via Nrf2-FXR-LXR pathway in NAFLD mice. Biomed Pharmacother. 105:274–281.
  • Yang Z-Z, Li L, Wang L, Yuan L-M, Xu M-C, Gu J-K, Jiang H-d, Yu L-S, Zeng S. 2017. The regioselective glucuronidation of morphine by dimerized human UGT2B7, 1A1, 1A9 and their allelic variants. Acta Pharmacol Sin. 38(8):1184–1194.
  • Yao H, Bai R, Ren T, Wang Y, Gu J, Guo Y. 2019. Enhanced platelet response to clopidogrel in Zucker diabetic fatty rats due to impaired clopidogrel inactivation by carboxylesterase 1 and increased exposure to active metabolite. Drug Metab Dispos. 47(8):794–801.
  • Yao H, Gu J, Shan Y, Wang Y, Chen X, Sun D, Guo Y. 2020. Type 2 diabetes mellitus decreases systemic exposure of clopidogrel active metabolite through upregulation of P-glycoprotein in rats. Biochem Pharmacol. 180:114142.
  • Yao X, Xia F, Tang W, Xiao C, Yang M, Zhou B. 2018. Isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics for the investigation of the effect of Hugan Qingzhi on non-alcoholic fatty liver disease in rats. J Ethnopharmacol. 212:208–215.
  • Yin H, Huang L, Ouyang T, Chen L. 2018. Baicalein improves liver inflammation in diabetic db/db mice by regulating HMGB1/TLR4/NF-κB signaling pathway. Int Immunopharmacol. 55:55–62.
  • Yin OQP, Lam SSL, Lo CMY, Chow MSS. 2004. Rapid determination of five probe drugs and their metabolites in human plasma and urine by liquid chromatography/tandem mass spectrometry: application to cytochrome P450 phenotyping studies. Rapid Commun Mass Spectrom. 18(23):2921–2933.
  • Yokota H, Iwano H, Endo M, Kobayashi T, Inoue H, Ikushiro S, Yuasa A. 1999. Glucuronidation of the environmental oestrogen bisphenol A by an isoform of UDP-glucuronosyltransferase, UGT2B1, in the rat liver. Biochem J. 340(2):405–409.
  • Yoon HY, Cho YA, Yee J, Gwak HS. 2020. Effects of CYP2B6 polymorphisms on plasma nevirapine concentrations: a systematic review and meta-analysis. Sci Rep. 10(1):17390.
  • Zhang C, Deng J, Liu D, Tuo X, Xiao L, Lai B, Yao Q, Liu J, Yang H, Wang N. 2018. Nuciferine ameliorates hepatic steatosis in high-fat diet/streptozocin-induced diabetic mice through a PPARα/PPARγ coactivator-1α pathway. Br J Pharmacol. 175(22):4218–4228.
  • Zhang L, Chu X, Wang H, Xie H, Guo C, Cao L, Zhou X, Wang G, Hao H. 2013. Dysregulations of UDP-glucuronosyltransferases in rats with valproic acid and high fat diet induced fatty liver. Eur J Pharmacol. 721(1-3):277–285.
  • Zhang L, Xu P, Cheng Y, Wang P, Ma X, Liu M, Wang X, Xu F. 2019. Diet-induced obese alters the expression and function of hepatic drug-metabolizing enzymes and transporters in rats. Biochem Pharmacol. 164:368–376.
  • Zhang M-H, Li J, Zhu X-Y, Zhang Y-Q, Ye S-T, Leng Y-R, Yang T, Zhang H, Kong L-Y. 2021. Physalin B ameliorates nonalcoholic steatohepatitis by stimulating autophagy and NRF2 activation mediated improvement in oxidative stress. Free Radic Biol Med. 164:1–12.
  • Zhang S, Song N, Li Q, Fan H, Liu C. 2008. Liquid chromatography/tandem mass spectrometry method for simultaneous evaluation of activities of five cytochrome P450s using a five-drug cocktail and application to cytochrome P450 phenotyping studies in rats. J Chromatogr B Analyt Technol Biomed Life Sci. 871(1):78–89.
  • Zhang Y, Hou K, Liu F, Luo X, He S, Hu L, Yang C, Huang L, Feng Y. 2021. The influence of CYP2C19 polymorphisms on voriconazole trough concentrations: systematic review and meta-analysis. Mycoses. 64(8):860–873.
  • Zhang Y, Wen J, Liu D, Qiu Z, Zhu Q, Li R, Zhang Y. 2021. Demethylenetetrahydroberberine alleviates nonalcoholic fatty liver disease by inhibiting the NLRP3 inflammasome and oxidative stress in mice. Life Sci. 281:119778.
  • Zhao K, Ding M, Cao H, Cao Z-X. 2012. In-vitro metabolism of glycyrrhetinic acid by human and rat liver microsomes and its interactions with six CYP substrates. J Pharm Pharmacol. 64(10):1445–1451.
  • Zhao M, Ma J, Li M, Zhang Y, Jiang B, Zhao X, Huai C, Shen L, Zhang N, He L, et al. 2021. Cytochrome P450 enzymes and drug metabolism in humans. Int J Mol Sci. 22(23). DOI:10.3390/ijms222312808
  • Zheng W, Song Z, Li S, Hu M, Shaukat H, Qin H. 2021. Protective effects of sesamol against liver oxidative stress and inflammation in high-fat diet-induced hepatic steatosis. Nutrients. 13(12):4484.
  • Zheng Z-G, Zhu S-T, Cheng H-M, Zhang X, Cheng G, Thu PM, Wang SP, Li H-J, Ding M, Qiang L, et al. 2021. Discovery of a potent SCAP degrader that ameliorates HFD-induced obesity, hyperlipidemia and insulin resistance via an autophagy-independent lysosomal pathway. Autophagy. 17(7):1592–1613.
  • Zhong X, Liu H. 2018. Honokiol attenuates diet-induced non-alcoholic steatohepatitis by regulating macrophage polarization through activating peroxisome proliferator-activated receptor γ. J Gastroenterol Hepatol. 33(2):524–532.
  • Zhou J, Zhou F, Wang W, Zhang X-J, Ji Y-X, Zhang P, She Z-G, Zhu L, Cai J, Li H. 2020. Epidemiological features of NAFLD from 1999 to 2018 in China. Hepatology. 71(5):1851–1864.
  • Zhou L, Sharma P, Yeo KR, Higashimori M, Xu H, Al-Huniti N, Zhou D. 2019. Assessing pharmacokinetic differences in Caucasian and East Asian (Japanese, Chinese and Korean) populations driven by CYP2C19 polymorphism using physiologically-based pharmacokinetic modelling. Eur J Pharm Sci. 139:105061.
  • Zhou W, Johnson TN, Bui KH, Cheung SYA, Li J, Xu H, Al-Huniti N, Zhou D. 2018. Predictive performance of physiologically based pharmacokinetic (PBPK) modeling of drugs extensively metabolized by major cytochrome P450s in children. Clin Pharmacol Ther. 104(1):188–200.
  • Zhou X, Li Z, Qi M, Zhao P, Duan Y, Yang G, Yuan L. 2020. Brown adipose tissue-derived exosomes mitigate the metabolic syndrome in high fat diet mice. Theranostics. 10(18):8197–8210.
  • Zou Y, Li J, Lu C, Wang J, Ge J, Huang Y, Zhang L, Wang Y. 2006. High-fat emulsion-induced rat model of nonalcoholic steatohepatitis. Life Sci. 79(11):1100–1107.