Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Drug Metabolism & Disposition
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Drug Metabolism & Disposition

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit dmd on Facebook
  • Follow dmd on Twitter
  • Follow ASPET on LinkedIn
Review ArticleSpecial Section on Drug Metabolism in Liver Injury and Repair

Withaferin A in the Treatment of Liver Diseases: Progress and Pharmacokinetic Insights

Yangliu Xia, Mingrui Yan, Ping Wang, Keisuke Hamada, Nana Yan, Haiping Hao, Frank J. Gonzalez and Tingting Yan
Drug Metabolism and Disposition May 2022, 50 (5) 685-693; DOI: https://doi.org/10.1124/dmd.121.000455
Yangliu Xia
School of Life Science and Medicine, Dalian University of Technology, Panjin, China (Y.X., M.Y.); Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Y.X., K.H., F.J.G., T.Y.); Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (P.W.); State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (N.Y., H.H.); and Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan (K.H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mingrui Yan
School of Life Science and Medicine, Dalian University of Technology, Panjin, China (Y.X., M.Y.); Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Y.X., K.H., F.J.G., T.Y.); Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (P.W.); State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (N.Y., H.H.); and Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan (K.H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ping Wang
School of Life Science and Medicine, Dalian University of Technology, Panjin, China (Y.X., M.Y.); Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Y.X., K.H., F.J.G., T.Y.); Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (P.W.); State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (N.Y., H.H.); and Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan (K.H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Keisuke Hamada
School of Life Science and Medicine, Dalian University of Technology, Panjin, China (Y.X., M.Y.); Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Y.X., K.H., F.J.G., T.Y.); Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (P.W.); State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (N.Y., H.H.); and Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan (K.H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nana Yan
School of Life Science and Medicine, Dalian University of Technology, Panjin, China (Y.X., M.Y.); Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Y.X., K.H., F.J.G., T.Y.); Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (P.W.); State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (N.Y., H.H.); and Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan (K.H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Haiping Hao
School of Life Science and Medicine, Dalian University of Technology, Panjin, China (Y.X., M.Y.); Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Y.X., K.H., F.J.G., T.Y.); Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (P.W.); State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (N.Y., H.H.); and Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan (K.H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Frank J. Gonzalez
School of Life Science and Medicine, Dalian University of Technology, Panjin, China (Y.X., M.Y.); Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Y.X., K.H., F.J.G., T.Y.); Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (P.W.); State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (N.Y., H.H.); and Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan (K.H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tingting Yan
School of Life Science and Medicine, Dalian University of Technology, Panjin, China (Y.X., M.Y.); Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Y.X., K.H., F.J.G., T.Y.); Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (P.W.); State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China (N.Y., H.H.); and Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan (K.H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

Withaferin A (WA) is a natural steroidal compound used in Ayurvedic medicine in India and elsewhere. Although WA was used as an anticancer reagent for decades, its role in the treatment of liver diseases has only recently been experimentally explored. Here, the effects of WA in the treatment of liver injury, systematic inflammation, and liver cancer are reviewed, and the toxicity and metabolism of WA as well as pharmacological potentials of other extracts from Withania somnifera (W. somnifera) discussed. The pharmacokinetic behaviors of WA are summarized and pharmacokinetic insights into current progress and future opportunities are highlighted.

SIGNIFICANCE STATEMENT This review outlines the current experimental progress of Withaferin A (WA) hepatoprotective activities and highlights gaps in the field. This work also discusses the pharmacokinetics of WA that can be used to guide future studies for the possible treatment of liver diseases with this compound.

Traditional herbs have been key sources for the development of hepatoprotective drugs for decades (Harvey et al., 2015; Yan et al., 2016; Yan et al., 2018; Yan et al., 2020). Impressive examples are silymarin for antifibrotic treatment (Gillessen and Schmidt, 2020), licorice-derived glycyrrhizin to treat viral hepatitis (Li et al., 2019), and berberine to treat nonalcoholic fatty liver diseases (NAFLD) (Sun et al., 2017; Sun et al., 2021). Some traditional herbs as drugs in the clinic or as the dietary supplement in the market still lack fully developed experimental evidence. As a result, the concepts of “reverse pharmacology” (Surh, 2011) and “reverse pharmacokinetics” (Hao et al., 2014) were proposed to advance the preclinical studies for traditional herbs. When meeting the increasing pursuit of precision medicine in modern times, globalization of traditional herbs is still facing great challenges, and extensive studies to evaluate the pharmacological effects of traditional herbs in combination with pharmacokinetic studies are attractive and indispensable for drug discovery.

Withania Somnifera (Ashwagandha, W. somnifera), belonging to the family Solanaceae, is a traditional herbal plant used as medicine or dietary supplement for decreasing inflammation; increasing energy; improving cognitive health, anxiety, and depression; as well as improving the homeostasis of glucose and cortisol in India. It has been used from the time of Ayurvedic and Unani systems of medicine at least since the 8th century (Mirjalili et al., 2009). Traditionally, the berries and leaves of W. somnifera were used as a local treatment of ulcers and tumors (Vanden Berghe et al., 2012). In addition, several reports have linked the health benefits of W. somnifera to its antidiabetic, antiepileptic, anti-inflammatory, antidepressant, and antiarthritic activities (Dutta et al., 2019; Huang et al., 2020). The clinical trials registered at clinicaltrials.gov using W. somnifera extract, also known as “ashwagandha” or “sensoril” as a dietary supplement or drug, are summarized in Table 1. Most of the pharmacological activities of W. somnifera have been attributed to the steroidal lactones and alkaloids called “withanolides,” among which the most important component is withaferin A (WA) (Wu et al., 2018; Huang et al., 2020).

View this table:
  • View inline
  • View popup
TABLE 1

Clinical trials for Withania somnifera registered in clinicaltrials.gov

WA was used as an anticancer reagent, and earlier reviews have documented the effects of WA in treating cancer (Vanden Berghe et al., 2012; Hassannia et al., 2020). Beyond its potential anticancer activities, the effects of WA in the treatment of liver diseases have only recently been studied. This review summarizes the main components in W. somnifera, the hepatoprotective effects and mechanisms of W. somnifera extracts, and WA for the treatment of various liver diseases, including acute liver injury, chronic liver injury, inflammation, and liver cancer.

Natural Products in W. Somnifera

More than 50 chemical constituents are contained in different parts of the root, leaf, fruit, and seed of W. somnifera (Kulkarni and Dhir, 2008). These chemical constituents include alkaloids, steroidal lactones, saponins with an additional acyl group, and withanolides that contain a glucose at carbon 27. Alkaloids and steroidal lactones were reported to be the main constituents of W. somnifera (Saleem et al., 2020). Although various alkaloids were found to be enriched in the root, withanine is the main constituent, with the other alkaloids, such as somniferine, somnine, somniferinine, and withananine, also detected. The leaves of W. somnifera consist primarily of steroidal lactones, which are commonly called “withanolide,” and are believed to account for its extraordinary medicinal properties (Sun et al., 2016). WA and withanolide D, were shown to have most of the pharmacological activity of W. somnifera (Sun et al., 2016). The major withanolides contained in W. somnifera are listed in Fig. 1.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Representative withanolides contained in W. somnifera. Compound 1, withaferin A; compound 2, withanone; compound 3, withanolide A; compounds 4–7, withanolide D–G; and compounds 8–12, withanolide I–M.

Hepatoprotective Effects of Natural Products in W. Somnifera

Effects of W. Somnifera Extracts in the Treatment of Liver Diseases

Several studies have evaluated the hepatoprotective potential of W. somnifera in animal models of various hepatic disorders. Aqueous root extract of W. somnifera administered at a dose of 500 mg/kg by gavage 60 minutes after acetaminophen (APAP) dosing was found to significantly reduce the elevated hepatotoxicity biomarkers, decrease lipid peroxidation, and enhance glutathione, catalase, glutathione reductase, and glutathione peroxidase activity in APAP-treated mice (Malik and Pandey, 2013). In another study, the ethanol extract of W. somnifera at 100 mg/kg protected against γ radiation-induced hepatotoxicity in rats; decreased serum alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and γ-glutamyl transpeptidase; decreased hepatic levels of malondialdehyde and total nitrate/nitrite; and increased hepatic antioxidant enzymes, including superoxide dismutase and glutathione peroxidase in rats (Hosny Mansour and Farouk Hafez, 2012). The hepatoprotective activity of a methanolic extract of W. somnifera roots was examined in APAP-intoxicated rats, and a significant hepatoprotective activity was observed when W. somnifera extract was given 2 hours prior to APAP administration through alleviating inflammatory and oxidative stress, accompanied by an inhibitory effect on hepatic levels of proinflammatory cytokines, including tumor necrosis factor α (TNFα), interleukin-1β (IL1β), cyclooxygenase 2, and inducible nitric oxide (Devkar et al., 2016). Among the components of W. somnifera extracts, WA was the most extensively studied among the natural compounds isolated from W. somnifera, whereas other abundant chemical constituents of W. somnifera have been studied to a much lesser extent in the treatment of liver diseases. Hence, in the following section of this review, a summary of the literature related to the effects of WA on the treatment of liver diseases is presented.

Effects of WA in the Treatment of Liver Injury

WA (Fig. 1, compound 3) was recognized as the most important bioactive component isolated from W. somnifera. Beyond the classic antitumor role of WA, a growing list of recent reports have shown that WA has activity against the obesity-associated metabolic syndromes (Lee et al., 2016), diabetes (Tekula et al., 2018), and systematic inflammation and liver disease (Gu et al., 2020). Although the detailed mechanisms by which WA can achieve these activities remains elusive, several hypotheses have been proposed, including leptin sensitizer for obesity treatment, immunoregulator for inflammation and fulminant acute liver injury, and activating antioxidant response for treatment of APAP-induced liver injury dependent on the NF-E2-related factor-2 (NRF2) or carbon tetrachloride (CCl4)-induced liver injury and fibrosis depending on sirtuin 3 (SIRT3). How WA may improve liver diseases are summarized in Fig. 2 for acute liver injury, Fig. 3 for chronic liver injury, and Fig. 4 for liver cancer.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

The role and mechanism of WA in the treatment of acute liver injury. In GalN/LPS-induced acute liver injury model, macrophage depletion using clodronate liposome were used to demonstrate that WA decreases the GalN/LPS-induced fulminant liver failure depending on its inhibitory effect toward macrophage activation, whereas global NLRP3 knockout mice (Nlrp3−/− mice) were used to demonstrate that WA decreased GalN/LPS-induced liver injury partially by inhibiting the NRLP3 inflammasome activation. In the APAP-induced acute liver injury model, WA attenuates APAP-induced hepatotoxicity accompanied by hepatic NRF2 activation. Although hepatocyte-specific Nrf2-null mice were used to determine the contribution of NRF2 to the hepatoprotective effect of WA in APAP model, whether the hepatopretecive effect of WA in treating APAP-induced liver injury is dependent on NRF2 signaling still warrants further study due to the questioned experimental process (dashed line).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

The role and mechanism of WA in the treatment of chronic liver injury. (A) In the HFD-induced obesity model, WA decreases obesity-associated metabolic syndrome and fatty liver as a leptin sensitizer. In the NASH model, WA both prevents and therapeutically alleviates the obese and lean NASH in accompanied by decreasing ceramides and ER stress. In HFD- and HFHC-fed ob/ob mice, WA therapeutically improves the obesity and NASH independent of the presence of leptin signaling. In ethanol-induced alcoholic liver injury model, WA therapeutically improves the alcoholic liver injury via inhibiting the hepatic lipogenesis. (B) In the non-NASH-associated fibrosis model induced by CCl4, bile duct ligation (BDL), or platelet-derived growth factor (PDGF), WA was demonstrated to activate SIRT3 to decrease the oxidative stress and liver fibrosis by using Sirt3−/− mice.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

The role and mechanism of WA in the treatment of liver cancer. In HCC, WA inhibits proliferation, migration, and invasion of HCC cells (Hep3B, HepG2, Huh7, QGY-7703, SK-Hep1, MHCC97H, and MHCC97L cells) by elevating the levels of p90-ribosomal S6 kinase, ETS-like transcription factor-1, DR5, and ERK and activating liver X receptor α to inhibit nuclear factor κB (NF-κB) transcriptional activity. In orthotopic liver tumors, WA inhibits liver tumor invasion and angiogenesis by downregulating the expression of pyruvate kinase (PYK2), rho-associated coiled-coil containing protein kinase 1 (ROCK1) and vascular endothelial growth factor (VEGF).

D-galactosamine/Lipopolysaccharide-Induced Fulminant Hepatitis

Fulminant hepatitis is a life-threatening clinical syndrome worldwide. WA was recently reported to attenuate D-galactosamine (GalN)/lipopolysaccharide (LPS)-induced hepatotoxicity in mice, associated with attenuating the inflammatory response via targeting macrophage and NLR family, pyrin domain containing 3 (NLRP3), while largely independent of NRF2 signaling, autophagy induction, and hepatic AMPKα1 and hepatocyte inhibitor of nuclear factor κB (Iκκβ) signaling (Xia et al., 2021). In this GalN/LPS-induced acute liver injury model, WA was found to have both potent preventive and therapeutic effects when mice were dosed with a single intraperitoneal injection of WA at 0.5 hour before or 2 hours after GalN dosing. By using clodronate liposome pretreatment to deplete macrophage, the hepatoprotective effect of WA was abolished, but by further using global Nlrp3-null mice as well as NLRP3-deficient primary macrophages, the WA effects were partially lost, but not totally abolished (Xia et al., 2021). In this same study, by using gene knockout mouse strains, the hepatoprotective effect of WA was found to be independent of the presence of hepatocyte AMPKα1, NRF2, and Iκκβ (Xia et al., 2021). Given that macrophage depletion abolished the hepatoprotective effects of WA in this model and NLRP3 knockout could not totally, but only partially, abolish the hepatoprotective effect of WA, additional targets located in macrophage, such as macrophage-specific Iκκβ, were suspected to mediate the hepatoprotective effects of WA in this model.

The GalN/LPS-induced acute liver injury model is characterized with systematic inflammation and hepatocyte apoptosis, whereas WA was found to have no direct role in TNFα-induced hepatocyte apoptosis (Xia et al., 2021). Decreasing LPS-induced systematic inflammation, also known as “cytokine storms” (Noori et al., 2020), was inferred to play a dominant role in the hepatoprotective effect of WA. Thus, with WA as a chemical probe, the results suggest that a strategy targeting the immune system, such as macrophage, to treat acute liver injury is achievable. This study suggests WA as a potential immunoregulator to normalize the altered immune response for the treatment of acute liver injury. Future studies on the effects of WA as one potent herbal immunoregulators in the treatment of other types of systematic inflammatory disorders are promising.

APAP-Induced Acute Liver Injury

WA was reported to protect against APAP-induced liver injury in two reports, indicating that NRF2 activation plays an important role in mediating the hepatocyte protective effects of WA (Jadeja et al., 2015; Palliyaguru et al., 2016). In an earlier study, a single dose of 40 mg/kg WA administered via intraperitoneal injection at 1 hour after APAP dosing significantly rescued APAP-induced hepatotoxicity, compared with the control vehicle ethanol (2 µL/g) (Jadeja et al., 2015), which suggests an efficient therapeutic effect. APAP-induced Jun N-terminal kinase (JNK) activation, mitochondrial B-cell lymphoma-2 (BCL-2)-associated X protein translocation, nitrotyrosine production, and hepatic inflammation were reduced by WA, accompanied by upregulation of NRF2 target genes, including Nrf2, GcLc, and Nqo1. In addition, WA alleviated H2O2-induced hepatocyte death and oxidative stress in the AML12 cell line, a nontransformed normal hepatocyte-derived cell line, in vitro (Jadeja et al., 2015). To better evaluate the therapeutic effect of one drug in the APAP model, APAP-treated mice are usually administered drugs for 2 hours or longer after APAP dosing to evaluate the effects on late-stage liver injury (Abdullah-Al-Shoeb et al., 2020). Comparing the hepatoprotective activity of a drug candidate to the clinical drug N-acetyl cysteine (Dear et al., 2021) could help to further assess the translational significance. Therefore, whether WA has a superior hepatoprotective effect against APAP-induced acute liver injury compared with N-acetyl cysteine, and whether WA still has a therapeutic effect at the later stages of APAP-induced liver injury warrant future study. In addition, this work only described the NRF2 activation effect of WA in APAP-treated mice (Jadeja et al., 2015); however, whether WA improvement of APAP-induced acute liver injury depends on its effect on NRF2 activation remains unexplored.

In a later study, WA was found to have an NRF2-dependent protective activity toward APAP-induced hepatotoxicity (Palliyaguru et al., 2016). WA exhibited direct NRF2-inducing activity both in mice in vivo and in mouse embryonic fibroblasts generated from fibroblasts isolated from 13.5-day-old mouse embryos in vitro. WA induced NRF2 signaling in a kelch like ECH associated protein 1 (KEAP1)-independent and phosphatase and tensin homolog (PTEN)/phosphatidylinositol 3-kinase (PI3K)/AKT serine/threonine kinase 1 (AKT1)-dependent manner in vitro as revealed by using NRF2/kelch like ECH associated protein 1 (KEAP1) double-knockout mouse embryonic fibroblasts (Palliyaguru et al., 2016). More importantly, this study employed a hepatocyte-specific Nrf2-null mouse line to confirm that the ameliorating effect of WA on APAP-induced liver injury was dependent on the presence of hepatocyte NRF2 (Palliyaguru et al., 2016), thereby supporting the contribution of WA-induced NRF2 signaling to the hepatoprotective effect of this compound in APAP-dosed mice. However, a major limitation of this study is that 100 µL of DMSO or 7 mg/kg of WA was administrated to mice via oral gavage 22 hours before APAP dosing. Given that WA showed a short retention time in vivo in previous pharmacokinetic studies (Thaiparambil et al., 2011; Vanden Berghe et al., 2012; Devkar et al., 2015; Dai et al., 2019), it is doubtful that WA could still achieve a hepatoprotective effect if only dosed once via gavage 22 hours before APAP, considering the anticipated low plasma concentration of this compound. DMSO is not suitable as a control vehicle for drugs when studying the APAP model, since DMSO has a hepatoprotective effect against APAP-induced liver injury even when used at a very low dose (Yoon et al., 2006). Thus, the dose and route of administration need to be considered when interpreting results. More studies are required to determine the extent by which NRF2 is involved in mitigating APAP toxicity by WA treatment.

Although the parent drug APAP is not hepatotoxic, APAP is metabolized into a toxic metabolite N-acetyl-p-benzoquinone (NAPQI) in the liver, a reaction catalyzed by cytochrome P450 (CYP) 2E1 (Lee et al., 1996; Cheung et al., 2005) and to a lesser degree by CYP3A11 and CYP1A2 (Zaher et al., 1998; Yan et al., 2016). Any factors that inhibit the generation of NAPQI could contribute to decreasing APAP-induced hepatotoxicity. How W. somnifera and WA affect APAP metabolism has not been examined in the APAP model in vivo and in vitro. However, several studies reported no significant inhibitory effects of W. somnifera extract on the CYPs that are involved in NAPQI generation from APAP (Savai et al., 2013; Varghese et al., 2014; Dey et al., 2015; Savai et al., 2015), indicating that it is less likely that WA exerts hepatoprotective effect toward APAP-induced liver injury by inhibiting CYP-mediated APAP metabolic activation. However, W. somnifera extract was found to exhibit inductive effects on CYP3A4 and CYP1A enzymes (Kumar et al., 2021). Thus, further studies are warranted to determine the extent by which metabolic activation of APAP is involved in reducing APAP-induced hepatotoxicity after W. somnifera and WA treatment.

Bromobenzene-Induced Hepatotoxicity

In an earlier study, a protective effect of W. somnifera extract was found in treating bromobenzene-induced nephrotoxicity and mitochondrial oxidative stress in rats through a proposed mechanism of decreasing mitochondrial oxidative stress (Vedi et al., 2014). More recently, pretreatment with WA provided a significant protection against bromobenzene-induced liver damage by preventing mitochondrial dysfunction through increasing activities of mitochondrial enzymes and balancing the expression of Bcl-2 associated X/Bcl-2 in liver (Vedi and Sabina, 2016). WA in W. somnifera at least partially contributes to the hepatoprotective effect of W. somnifera in this bromobenzene-induced liver injury model.

WA in the Metabolic Diseases and Nonalcoholic Fatty Liver Disease

As one of the most prevalent chronic liver diseases worldwide, nonalcoholic fatty liver disease (NAFLD) affects 20%–25% of the adult population (Cotter and Rinella, 2020). NAFLD is a typical type of obesity-associated metabolic syndrome, which is frequently is associated with, but is not limited to, obesity (Younes and Bugianesi, 2019; Cotter and Rinella, 2020). NAFLD includes simple nonalcoholic fatty liver (NAFL) characterized with hepatic steatosis and mild inflammation and the progressive nonalcoholic steatohepatitis (NASH) characterized by severe liver inflammation and fibrosis, which could further progress to end-stage liver diseases, including cirrhosis and hepatocellular carcinoma (HCC) (Schwabe et al., 2020; Sheka et al., 2020; Francque et al., 2021). The role of WA in treating diet-induced NAFL and progressive NASH is diagrammed in Fig. 3A.

An initial study demonstrated that WA alleviated obesity-associated metabolic syndrome and hepatic steatosis by acting as a leptin sensitizer (Lee et al., 2016). In this study, 12-week WA (1.25 mg/kg/d, i.p.) treatment reduced the levels of proinflammatory cytokines including toll-like receptor, nuclear factor κB (NFκB), tumor necrosis factor-α (TNFα), chemokine ligand-receptor, and cyclooxygenase 2 in both the serum and liver of high-fat diet (HFD)-fed obese mice, along with protective activity against obesity, oxidative stress, and insulin resistance (Lee et al., 2016). Moreover, WA (6 mg/kg, 3 times per week, i.p.) was found to therapeutically improve insulin sensitivity, downregulate the inflammatory response, and decrease body weight and hepatic steatosis in established HFD-fed obese mice, which was suggested to be partly through upregulating peroxisome proliferator-activated receptor gamma phosphorylation-related downstream genes, including carbonic anhydrase 3, selenium binding protein 1, amyloid beta precursor-like protein 2, thioredoxin interacting protein, and adiponectin (Khalilpourfarshbafi et al., 2019). The antihepatic inflammation effect of WA was also observed and described in pre-existing HFD-induced obese mice in a therapeutic manner when mice were fed a HFD for 12 weeks and then dosed by oral gavage with WA at 1.25 mg/kg/d for 12 weeks (Abu Bakar et al., 2019). These studies support the view that WA has a therapeutic effect in treating obesity-induced metabolic syndrome that includes hepatic steatosis, inflammation, and diabetes. However, these HFD-induced obesity models may only represent the mild fatty liver phenotype, NAFL, while still at the early stage of NAFLD.

The effect of WA on the later stages of NAFLD, NASH, has also been examined. WA was demonstrated to have a potent therapeutic effect in decreasing NASH-associated symptoms in both obese and lean NASH models. In several widely used NASH models, WA had both preventive and therapeutic effects in improving NASH, with reduced serum aminotransferase levels, liver steatosis, inflammation, endoplasmic reticulum (ER) stress, and fibrosis in the WA-treated group compared with the vehicle-treated group, and this was correlated with normalized serum ceramides, hepatic ceramide catabolism, and ER stress (Patel et al., 2019). In this study, methionine-choline-deficient diet-fed wild-type C57BL/6N mice were used to induce a lean NASH model, the 40% high-fat and high-cholesterol (HFHC) diet-fed wild-type C57BL/6N mice were used to establish an obese NASH model, whereas 40% HFHC-fed ob/ob mice were employed to generate a leptin signaling-deficient NASH model (Patel et al., 2019). Beyond the preventive effects, WA improved all NASH symptoms in a therapeutic manner when dosed at the later stage of the established NASH models. Given that WA was found to improve methionine-choline-deficient-induced NASH, a lean NASH model, the hepatoprotective effect of WA against NASH was inferred to be at least partially independent of its antiobesity effect. Using ob/ob mice, WA was confirmed to produce an anti-NASH effect independent of leptin signaling (Patel et al., 2019).

WA in Non-NASH-Associated Liver Fibrosis

The effect of WA in non-NASH-associated fibrosis are summarized in Fig. 3B. In a bile duct ligation-induced liver fibrosis model, WA inhibited epithelial mesenchymal transition process by inhibiting the expression of enzymes such as MMP2, TIMP1, and LOXL2, and the transcriptional repressor SNAIL1, thus enhancing the expression of CDH1 leading to the reversal of epithelial mesenchymal transition (Sayed et al., 2019). In the liver fibrosis models induced by platelet-derived growth factor BB and CCl4, WA attenuated liver fibrosis by inhibiting oxidative stress in a SIRT3-dependent manner as revealed by using SIRT3 knockout mice and Sirt3 silencing in JS1 cells, an immortalized mouse hepatic stellate cell line (Gu et al., 2020). These studies further extend the antifibrosis scope of WA in different types of hepatic fibrosis models. The direct effects and detailed mechanisms of WA in alleviating hepatic fibrogenesis, such as hepatic stellate activation, macrophage activation, and hepatocyte damage, warrant additional studies.

WA in Alcoholic Liver Injury

The hepatoprotective effect of WA in alcoholic liver injury was investigated in an acute-upon-chronic liver injury model (Hamada et al., 2022). In this binge model, WA significantly decreased binge ethanol-induced liver injury and hepatic lipid accumulation both in a preventive and therapeutic manner. Mechanistically, WA inhibits ethanol-induced lipogenesis in vivo as well as in primary hepatocytes in vitro. This study suggests the therapeutic potential of WA in the treatment of alcoholic liver injury (Hamada et al., 2022). The direct target of WA and the role of WA in long-term alcohol-induced alcoholic steatohepatitis and fibrosis warrants further study.

WA in Liver Cancer

The effect of WA on tumor growth and metastasis in liver was investigated in a nude mouse model. WA significantly decreased tumor growth, the incidence of lung metastasis, and macrophage infiltration in liver tumors and vessels by decreasing cell migration and tumor vascular endothelial cell damage, and inducing tumor necrosis through inhibiting the expression of pyruvate kinase PYK2, rho-associated coiled-coil containing protein kinase 1 and vascular endothelial growth factor (Wang et al., 2015). In another study, WA was found to suppress the proliferation, migration, invasion, and anchorage-independent growth of HCC by inhibiting NFκB signaling through liver X receptor α activation in HCC cells, including Hep3B, HepG2, Huh7, and QGY-7703 cells (Shiragannavar et al., 2021). In addition, WA sensitized tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis in several human HCC cell lines, including Huh7, SK-Hep1, and Hep3B cells, by death receptor protein-5 (DR5) upregulation and CASP8 and FADD like apoptosis regulator downregulation (Lee et al., 2009). In addition to inducing apoptotic cell death, the WA inhibitory effect on the growth of HCC cells, including Huh7, HepG2, MHCC97H, and MHCC97L, was diminished by the concomitant induction of autophagy, which could be rescued by cotreatment with chloroquine, an autophagy inhibitor (Siddharth et al., 2019). Administration of WA was also reported to decrease the level of serum tumor marker α-fetoprotein and rescue the diethylnitrosamine-induced HCC in rats (Murugan et al., 2015). Oral administration of WA effectively inhibited HepG2-xenograft growth and diethylnitrosamine-induced HCC by concomitantly increasing p90-ribosomal S6 kinase phosphorylation and activating ETS-like transcription factor-1 and DR5 through increasing phosphorylation of extracellular signal-regulated kinases (ERK) (Kuppusamy et al., 2017). The roles of WA in the treatment of liver tumor are summarized in Fig. 4.

WA in Systematic Inflammation

Although inhibiting inflammation potentially decreases liver injury (Zhang et al., 2019), WA was reported to have potent anti-inflammation activities. WA inhibited the expression of inducible nitric oxide at both the protein and mRNA levels in LPS-stimulated RAW264.7 cells, a macrophage-like, Abelson leukemia virus-transformed cell line derived from BALB/c mice, by blocking the phosphorylation of AKT serine/threonine kinase 1 9AKT1) and ERK via inhibition of inhibitor of NFκB (IκB) phosphorylation and subsequent NFκB activation (Oh et al., 2008). WA was found to exert its anti-inflammatory effects by inhibiting NFκB activation through targeting Iκκβ in HEK293T cells, a cell line derived from human embryonic kidney cells (Heyninck et al., 2014). In another study, WA inhibited inflammation by potently inhibiting TNFα-induced NFκB activation via inhibition of Iκκβ kinase activity in vitro in the mouse fibrosarcoma cell line L929sA and human embryonic kidney 293T cells, and in an acute inflammatory mouse model in vivo (Kaileh et al., 2007). WA dose-dependently inhibited the IL1β secretion as well as the cleavage of IL1β and caspase 1 in LPS-primed macrophage via inhibiting NLRP3 inflammasome activation (Kim et al., 2015). WA also reduced the release of IL1β and IL18 via inhibition of NFκB activity and suppression of NLRP3 inflammasome activation in THP-1 cells (Dubey et al., 2018).

Hepatoprotective and Anti-Inflammation Potential of Other Extracts

Both WA and another withanolide constituent from W. somnifera, withanone, significantly decreased the production of proinflammatory cytokines TNFα and IL6 at both the mRNA and protein levels in LPS-stimulated bone marrow-derived macrophages (Purushotham et al., 2017). Further mechanistic studies showed that WA markedly inhibited the mitogen-activated protein kinase. including ERK, JNK, and p38 as well as NFκB activation, whereas withanone regulated only ERK and JNK signaling pathways (Purushotham et al., 2017). One withanolide constituent protected liver cells from oxidative stress-induced damage by increasing cellular survival rate and activities of superoxide dismutase, glutathione, and catalase and reducing the accumulation of reactive oxygen species via activating nuclear translocation of NRF2 and increasing expression of the NRF2 target gene heme oxygenase 1 (Wang et al., 2018). Six withanolides showed anti-inflammatory activities through inhibition of nitric oxide production in murine macrophage RAW 264.7 cells stimulated by LPS (Wu et al., 2018). Withanolide A inhibited oxidative and inflammation in the immortalized mouse microglial cell line BV-2 via simultaneously suppressing the NFκB pathways, inhibiting LPS-induced nitric oxide production, and stimulating the NRF2/heme oxygenase 1 pathway, but this antioxidative and anti-inflammatory effect was 10-fold less efficacious than WA (Sun et al., 2016).

Toxicity of WA

The toxicity of WA was investigated both in animal models and in humans by using W. somnifera extracts standardized for WA or pure WA. An oral LD50 of W. somnifera extracts in rats was greater than 2000 mg/kg body weight in acute toxicity studies (Patel et al., 2016). Further, a subacute toxicity study also showed that rats administered W. somnifera extracts for 28 days did not exhibit any toxicologically significant changes in the brain, liver, and kidney compared with the control group (Patel et al., 2016). A safety evaluation of W. somnifera extracts standardized for WA in a phase I trial demonstrated that the capsule formulation of WA comprised of 72, 108, 144, and 216 mg of WA administered in two or four divided doses per day for at least 30 days was generally well-tolerated with no severe adverse events, but slight adverse effects, such as elevation of liver enzymes (5/11) and skin rash (2/11), were observed (Pires et al., 2020). Toxicity studies with pure WA has not been reported, and only two studies suggested that WA was toxic to mice with an LD50 of 54 mg/kg body weight, which is much higher than the pharmacological doses typically employed that range between 1.25–10 mg/kg (Shohat et al., 1967; Shohat et al., 1970; Lee et al., 2016; Patel et al., 2019; Xia et al., 2021). The toxicity between pure WA and plant extracts of W. somnifera could differ because of purity and composition concerns, which is an issue that demands future study.

Metabolism of WA

Metabolism of WA

Studies on the pharmacokinetics and bioavailability of WA are summarized in Fig. 5 and Table 2. WA was found to be impermeable in an in vitro absorption model using MDCK cells derived from canine kidney (Devkar et al., 2015). WA is readily transported across Caco‐2 cell plasma membranes in vitro, whereas WA had an oral bioavailability of 32.4 ± 4.8% in vivo, based on the studies of intravenous (5 mg/kg) and oral (10 mg/kg) dosing in male rats (Dai et al., 2019). Extensive first-pass metabolism of WA was further suggested by rat intestine‐liver in situ perfusion, where WA concentration in the media was rapidly decreased and only 27.1% remained within 1 hour (Dai et al., 2019). Pharmacokinetic studies on WA showed a rapid plasma clearance in mice administered a single dose of WA via intraperitoneal injection (Thaiparambil et al., 2011; Vanden Berghe, 2012). A study reporting pharmacokinetic evaluation of WA after oral administration demonstrated a short half-life with the value of 59.9 minutes for this constituent in mice administered a single oral dose of W. somnifera extracts (Patil et al., 2013). A previous report found a relatively short half-life (t1/2 = 2.0 hours) of WA in mice after a single intraperitoneal injection of 5 mg/kg WA (Patel et al., 2019). Moreover, the observation that no detectable WA was found in plasma samples in humans orally administered a capsule formulation of WA at 72–216 mg/kg in two or four divided doses per day for at least 30 days as measured by high performance liquid chromatography with a limit of quantitation of 50 ng/mL (Pires et al., 2020). However, few studies evaluated the metabolic route of WA. Others found that WA was metabolized via hydroxylation, hydrogenation, and hydrolysis (Rosazza et al., 1978; Funska et al., 1985; Fuska et al., 1987), which was further supported by a more recent study where metabolites of WA generated from these pathways were found in rats (Dai et al., 2019). Due to the structural similarities of steroidal lactone classification between WA and endogenous steroids, it likely undergoes oxidative metabolism mediated by CYPs. More detailed future investigations are needed to characterize the metabolic pathway(s) as well as the major metabolizing enzymes responsible for WA metabolism in vivo.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

The proposed metabolism routes of WA. Seven metabolites, M1–M7, were identified via hydroxylation, hydrogenation, and hydrolysis of WA in rat or human liver microsomes, with M2, M3, and M7 specifically found in human liver microsomes.

View this table:
  • View inline
  • View popup
TABLE 2

The pharmacokinetic parameters of WA in experimental animals

Pharmacokinetic Insights into Future Directions

Poor pharmacokinetic behavior indicated that the hepatoprotective activity of WA may be attributed to its metabolites. Phase I reactions convert a parent drug to metabolites that, in some cases, could possess superior efficacy and be more biologically active and water-soluble than the parent compound by unmasking or inserting a polar functional group. The generated active metabolites, as “metabolized” molecules, can be less prone to first pass metabolism and thus have improved pharmacokinetic behaviors compared with the parent drugs (Sun and Wesolowski, 2021). Therefore, it is of great interest to compare the hepatoprotective effects of WA and its metabolites side by side to elucidate the effective substance basis of WA and explore new drug candidates in treating liver diseases.

On the other hand, it is also worth noting that decreased expression levels of phase I and II enzymes and drug transporters are often observed under pathologic states of the liver, including acute liver injury, the metabolic diseases and NAFLD, liver fibrosis, liver cancer, and systematic inflammation (Hanada et al., 2012; Wu and Lin, 2019; Bao et al., 2020; Feng et al., 2020), which may lead to reduced metabolism of WA. Thus, it is important that pharmacologists keep this in mind for designing appropriate doses of WA when using liver injury models. Whether WA is toxic is still controversial, and more toxicological evaluation is needed to determine the safety of WA. In addition, different formulations may need to be studied so that the adjusted pharmacokinetic properties (absorption, distribution, metabolism, and elimination) will yield optimal efficacy. Indeed, WA embedded in polycaprolactone implants was proposed to overcome the problems regarding its bioavailability and pharmacokinetics (Gupta et al., 2012), thereby allowing long-term systemic circulation for controlled treatment.

The exact mechanism by which WA alleviated liver diseases remains largely unknown, due in part to that fact that no receptor for this compound has been identified. Regarding the potentially extensive metabolism of WA in vivo, it is not clear whether WA, the parent drug, or its biologically active metabolite(s) may directly contribute to its hepatoprotective effects. Due to the short half-life of WA after gavage in vivo, the pharmacological effects achieved after oral intake of WA may be due in part to modulation of the host intestinal targets, gut microbiota, or other factors involved in the gut-liver axis. The specific intestinal targets with which WA interacts to produce its pharmacological effects on liver diseases after oral gavage warrant further investigation. Various methodologies, including such as RNA sequencing, proteomics, metagenomics, and untargeted metabolomics, are needed to flush out mechanistic clues. Pharmacokinetic studies are also needed to be performed to clarify the distribution of WA and its potential active metabolites, which could direct future mechanistic research to better select a target organ/cell for analyses.

Conclusion

This review summarizes WA therapeutic potential in liver diseases, including acute liver injury, the metabolic diseases and NAFLD, liver fibrosis, liver cancer, and systematic inflammation. WA exerts its hepatoprotective effects mainly through antioxidant and anti-inflammatory activities in part by modulating NRF2 and NFκB signaling and the NLRP3 inflammasome. The antitumor effects are efficient at the safe doses of WA employed in rodent models, suggesting a selectivity of WA toward killing liver tumor cells compared with its activity in normal hepatocytes. WA is known to protect against liver cancer both in rodent models and in HCC cell lines in vitro.

The pharmacological and toxicological properties of WA, especially in humans, have been poorly studied. Structure modifications to make it circulate longer and yet retain its biologic activity may be of value in drug discovery. However, it should be noted that WA is found to be absorbed at low levels in WA-containing natural products used to treat humans, and thus the potential hepatoprotective effects of WA metabolites as well as the other components deserve further study. WA holds a great potential for novel drug discovery in the treatment of both acute liver injury, chronic liver injury, metabolic diseases as well as liver tumor, which deserves extensive future studies.

Acknowledgments

The authors thank Diane Cooper, MSLS, NIH Library, for editing the manuscript.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Xia, M. Yan, Wang, Hamada, N. Yan, Hao, Gonzalez, T. Yan.

Footnotes

    • Received March 9, 2021.
    • Accepted December 6, 2021.
  • This work was supported by National Key Research and Development Program of China [Grant 2021YFE0200900] (to P.W.), the Fundamental Research Funds for the Central Universities [Grant DUT20RC (4) 013] (to Y.X. and M.Y.), and National Institutes of Health Center for Cancer Research, National Cancer Institute Intramural Research Program.

  • No author has an actual or perceived conflict of interest with the contents of this article.

  • https://dx.doi.org/10.1124/dmd.121.000455.

Abbreviations

 APAP
acetaminophen
BCL-2
B-cell lymphoma-2
CCl4
carbon tetrachloride
CYP
cytochrome P450;
DR5
death receptor protein-5
ER
endoplasmic reticulum
ERK
extracellular-signal-regulated kinase
GalN
D-galactosamine
HCC
hepatocellular carcinoma
HFD
high-fat diet
HFHC
high-fat and high-cholesterol
Iκκβ
inhibitor of nuclear factor κB
IL1β
interleukin 1β
JNK
Jun N-terminal kinase
LPS
lipopolysaccharide
NAFL
nonalcoholic fatty liver
NAFLD
nonalcoholic fatty liver disease
NAPQI
N-acetyl-p-benzoquinone
NASH
nonalcoholic steatohepatitis
NFκB
nuclear factor κB
NLRP3
NLR family, pyrin domain containing 3
NRF2
NF-E2-related factor-2
SIRT3
sirtuin 3
TNFα
tumor necrosis factor α
WA
withaferin A
W. somnifera
Withania somnifera
  • U.S. Government work not protected by U.S. copyright

References

  1. ↵
    1. Abdullah-Al-Shoeb M,
    2. Sasaki K,
    3. Kikutani S,
    4. Namba N,
    5. Ueno K,
    6. Kondo Y,
    7. Maeda H,
    8. Maruyama T,
    9. Irie T, and
    10. Ishitsuka Y
    (2020) The late-stage protective effect of mito-TEMPO against acetaminophen-induced hepatotoxicity in mouse and three-dimensional cell culture models. Antioxidants 9:965.
    OpenUrl
  2. ↵
    1. Abu Bakar MH,
    2. Azmi MN,
    3. Shariff KA, and
    4. Tan JS
    (2019) Withaferin A protects against high-fat diet-induced obesity via attenuation of oxidative stress, inflammation, and insulin resistance. Appl Biochem Biotechnol 188:241–259.
    OpenUrl
  3. ↵
    1. Bao Y,
    2. Wang P,
    3. Shao X,
    4. Zhu J,
    5. Xiao J,
    6. Shi J,
    7. Zhang L,
    8. Zhu HJ,
    9. Ma X,
    10. Manautou JE, et al.
    (2020) Acetaminophen-induced liver injury alters expression and activities of cytochrome P450 enzymes in an age-dependent manner in mouse liver. Drug Metab Dispos 48:326–336.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Cheung C,
    2. Yu AM,
    3. Ward JM,
    4. Krausz KW,
    5. Akiyama TE,
    6. Feigenbaum L, and
    7. Gonzalez FJ
    (2005) The cyp2e1-humanized transgenic mouse: role of cyp2e1 in acetaminophen hepatotoxicity. Drug Metab Dispos 33:449–457.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Cotter TG and
    2. Rinella M
    (2020) Nonalcoholic fatty liver disease 2020: the state of the disease. Gastroenterology 158:1851–1864.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Dai T,
    2. Jiang W,
    3. Guo Z,
    4. Wang Z,
    5. Huang M,
    6. Zhong G,
    7. Liang C,
    8. Pei X, and
    9. Dai R
    (2019) Studies on oral bioavailability and first-pass metabolism of withaferin A in rats using LC-MS/MS and Q-TRAP. Biomed Chromatogr 33:e4573.
    OpenUrl
  7. ↵
    1. Dear JW,
    2. Ng ML,
    3. Bateman DN,
    4. Leroy Sivappiragasam P,
    5. Choi H,
    6. Khoo BBJ,
    7. Ibrahim B, and
    8. Drum CL
    (2021) A metabolomic analysis of thiol response for standard and modified N-acetyl cysteine treatment regimens in patients with acetaminophen overdose. Clin Transl Sci 14:1476–1489.
    OpenUrl
  8. ↵
    1. Devkar ST,
    2. Kandhare AD,
    3. Sloley BD,
    4. Jagtap SD,
    5. Lin J,
    6. Tam YK,
    7. Katyare SS,
    8. Bodhankar SL, and
    9. Hegde MV
    (2015) Evaluation of the bioavailability of major withanolides of Withania somnifera using an in vitro absorption model system. J Adv Pharm Technol Res 6:159–164.
    OpenUrl
  9. ↵
    1. Devkar ST,
    2. Kandhare AD,
    3. Zanwar AA,
    4. Jagtap SD,
    5. Katyare SS,
    6. Bodhankar SL, and
    7. Hegde MV
    (2016) Hepatoprotective effect of withanolide-rich fraction in acetaminophen-intoxicated rat: decisive role of TNF-α, IL-1β, COX-II and iNOS. Pharm Biol 54:2394–2403.
    OpenUrl
  10. ↵
    1. Dey D,
    2. Chaskar S,
    3. Athavale N, and
    4. Chitre D
    (2015) Acute and chronic toxicity, cytochrome p450 enzyme inhibition, and HERG channel blockade studies with a polyherbal, ayurvedic formulation for inflammation. BioMed Res Int 2015:971982.
    OpenUrl
  11. ↵
    1. Dubey S,
    2. Yoon H,
    3. Cohen MS,
    4. Nagarkatti P,
    5. Nagarkatti M, and
    6. Karan D
    (2018) Withaferin A associated differential regulation of inflammatory cytokines. Front Immunol 9:195.
    OpenUrl
  12. ↵
    1. Dutta R,
    2. Khalil R,
    3. Green R,
    4. Mohapatra SS, and
    5. Mohapatra S
    (2019) Withania somnifera (ashwagandha) and withaferin A: potential in integrative oncology. Int J Mol Sci 20:5310.
    OpenUrl
  13. ↵
    1. Feng S,
    2. Li A,
    3. Zheng YC, and
    4. Liu HM
    (2020) Role of drug-metabolizing enzymes in cancer and cancer therapy. Curr Drug Metab 21:67–76.
    OpenUrl
  14. ↵
    1. Francque S,
    2. Szabo G,
    3. Abdelmalek MF,
    4. Byrne CD,
    5. Cusi K,
    6. Dufour JF,
    7. Roden M,
    8. Sacks F, and
    9. Tacke F
    (2021) Nonalcoholic steatohepatitis: the role of peroxisome proliferator-activated receptors. Nat Rev Gastroenterol Hepatol 18:24–39.
    OpenUrlCrossRef
  15. ↵
    1. Funska J,
    2. Khandlová A,
    3. Šturdíková M,
    4. Rosazza JP, and
    5. Kieslich K
    (1985) Biotransformation of withaferin-A by a culture of arthrobacter simplex. Folia Microbiol 30:427–432.
    OpenUrl
  16. ↵
    1. Fuska J,
    2. Proska B,
    3. Williamson J, and
    4. Rosazza JP
    (1987) Microbiological and chemical dehydrogenation of withaferin A. Folia Microbiol (Praha) 32:112–115.
    OpenUrlPubMed
  17. ↵
    1. Gillessen A and
    2. Schmidt HH
    (2020) Silymarin as supportive treatment in liver diseases: a narrative review. Adv Ther 37:1279–1301.
    OpenUrl
  18. ↵
    1. Gu J,
    2. Chen C,
    3. Wang J,
    4. Chen T,
    5. Yao W,
    6. Yan T, and
    7. Liu Z
    (2020) Withaferin A exerts preventive effect on liver fibrosis through oxidative stress inhibition in a sirtuin 3-dependent manner. Oxid Med Cell Longev 2020:2452848.
    OpenUrl
  19. ↵
    1. Gupta RC,
    2. Bansal SS,
    3. Aqil F,
    4. Jeyabalan J,
    5. Cao P,
    6. Kausar H,
    7. Russell GK,
    8. Munagala R,
    9. Ravoori S, and
    10. Vadhanam MV
    (2012) Controlled-release systemic delivery—a new concept in cancer chemoprevention. Carcinogenesis 33:1608–1615.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Hamada K,
    2. Wang P,
    3. Xia Y,
    4. Yan N,
    5. Takahashi S,
    6. Krausz KW,
    7. Hao H,
    8. Yan T, and
    9. Gonzalez FJ
    (2022) Withaferin A alleviates ethanol-induced liver injury by inhibiting hepatic lipogenesis. Food Chem Toxicol 160:112807.
    OpenUrl
  21. ↵
    1. Hanada K,
    2. Nakai K,
    3. Tanaka H,
    4. Suzuki F,
    5. Kumada H,
    6. Ohno Y,
    7. Ozawa S, and
    8. Ogata H
    (2012) Effect of nuclear receptor downregulation on hepatic expression of cytochrome P450 and transporters in chronic hepatitis C in association with fibrosis development. Drug Metab Pharmacokinet 27:301–306.
    OpenUrlPubMed
  22. ↵
    1. Hao H,
    2. Zheng X, and
    3. Wang G
    (2014) Insights into drug discovery from natural medicines using reverse pharmacokinetics. Trends Pharmacol Sci 35:168–177.
    OpenUrlCrossRef
  23. ↵
    1. Harvey AL,
    2. Edrada-Ebel R, and
    3. Quinn RJ
    (2015) The re-emergence of natural products for drug discovery in the genomics era. Nat Rev Drug Discov 14:111–129.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Hassannia B,
    2. Logie E,
    3. Vandenabeele P,
    4. Vanden Berghe T, and
    5. Vanden Berghe W
    (2020) Withaferin A: from ayurvedic folk medicine to preclinical anti-cancer drug. Biochem Pharmacol 173:113602.
    OpenUrl
  25. ↵
    1. Heyninck K,
    2. Lahtela-Kakkonen M,
    3. Van der Veken P,
    4. Haegeman G, and
    5. Vanden Berghe W
    (2014) Withaferin A inhibits NF-kappaB activation by targeting cysteine 179 in IKKβ. Biochem Pharmacol 91:501–509.
    OpenUrlCrossRefPubMed
  26. ↵
    1. Hosny Mansour H and
    2. Farouk Hafez H
    (2012) Protective effect of Withania somnifera against radiation-induced hepatotoxicity in rats. Ecotoxicol Environ Saf 80:14–19.
    OpenUrlPubMed
  27. ↵
    1. Huang M,
    2. He JX,
    3. Hu HX,
    4. Zhang K,
    5. Wang XN,
    6. Zhao BB,
    7. Lou HX,
    8. Ren DM, and
    9. Shen T
    (2020) Withanolides from the genus physalis: a review on their phytochemical and pharmacological aspects. J Pharm Pharmacol 72:649–669.
    OpenUrl
  28. ↵
    1. Jadeja RN,
    2. Urrunaga NH,
    3. Dash S,
    4. Khurana S, and
    5. Saxena NK
    (2015) Withaferin-A reduces acetaminophen-induced liver injury in mice. Biochem Pharmacol 97:122–132.
    OpenUrl
  29. ↵
    1. Kaileh M,
    2. Vanden Berghe W,
    3. Heyerick A,
    4. Horion J,
    5. Piette J,
    6. Libert C,
    7. De Keukeleire D,
    8. Essawi T, and
    9. Haegeman G
    (2007) Withaferin A strongly elicits IkappaB kinase beta hyperphosphorylation concomitant with potent inhibition of its kinase activity. J Biol Chem 282:4253–4264.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Khalilpourfarshbafi M,
    2. Devi Murugan D,
    3. Abdul Sattar MZ,
    4. Sucedaram Y, and
    5. Abdullah NA
    (2019) Withaferin A inhibits adipogenesis in 3T3-F442A cell line, improves insulin sensitivity and promotes weight loss in high fat diet-induced obese mice. PLoS One 14:e0218792.
    OpenUrl
  31. ↵
    1. Kim JE,
    2. Lee JY,
    3. Kang MJ,
    4. Jeong YJ,
    5. Choi JA,
    6. Oh SM,
    7. Lee KB, and
    8. Park JH
    (2015) Withaferin A inhibits helicobacter pylori-induced production of IL-1β in dendritic cells by regulating NF-κB and NLRP3 inflammasome activation. Immune Netw 15:269–277.
    OpenUrl
  32. ↵
    1. Kulkarni SK and
    2. Dhir A
    (2008) Withania somnifera: an Indian ginseng. Prog Neuropsychopharmacol Biol Psychiatry 32:1093–1105.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Kumar S,
    2. Bouic PJ, and
    3. Rosenkranz B
    (2021) Investigation of CYP2B6, 3A4 and β-esterase interactions of Withania somnifera (L.) dunal in human liver microsomes and HepG2 cells. J Ethnopharmacol 270:113766.
    OpenUrl
  34. ↵
    1. Kuppusamy P,
    2. Nagalingam A,
    3. Muniraj N,
    4. Saxena NK, and
    5. Sharma D
    (2017) Concomitant activation of ETS-like transcription factor-1 and death receptor-5 via extracellular signal-regulated kinase in withaferin A-mediated inhibition of hepatocarcinogenesis in mice. Sci Rep 7:17943.
    OpenUrl
  35. ↵
    1. Lee J,
    2. Liu J,
    3. Feng X,
    4. Salazar Hernández MA,
    5. Mucka P,
    6. Ibi D,
    7. Choi JW, and
    8. Ozcan U
    (2016) Withaferin A is a leptin sensitizer with strong antidiabetic properties in mice. Nat Med 22:1023–1032.
    OpenUrl
  36. ↵
    1. Lee SS,
    2. Buters JT,
    3. Pineau T,
    4. Fernandez-Salguero P, and
    5. Gonzalez FJ
    (1996) Role of CYP2E1 in the hepatotoxicity of acetaminophen. J Biol Chem 271:12063–12067.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    1. Lee T-J,
    2. Um HJ,
    3. Min S,
    4. Park J-W,
    5. Choi KS, and
    6. Kwon TK
    (2009) Withaferin A sensitizes TRAIL-induced apoptosis through reactive oxygen species-mediated up-regulation of death receptor 5 and down-regulation of c-FLIP. Free Radic Biol Med 46:1639–1649.
    OpenUrlCrossRefPubMed
  38. ↵
    1. Li X,
    2. Sun R, and
    3. Liu R
    (2019) Natural products in licorice for the therapy of liver diseases: progress and future opportunities. Pharmacol Res 144:210–226.
    OpenUrl
  39. ↵
    1. Malik T and
    2. Pandey DK
    (2013) Ameliorative potential of aqueous root extract of Withania somnifera against paracetamol induced liver damage in mice. Pharmacologia 4:89–94.
    OpenUrl
  40. ↵
    1. Mirjalili MH,
    2. Moyano E,
    3. Bonfill M,
    4. Cusido RM, and
    5. Palazón J
    (2009) Steroidal lactones from Withania somnifera, an ancient plant for novel medicine. Molecules 14:2373–2393.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Murugan S,
    2. Madhusoodanan N,
    3. Shankar S, and
    4. Babu V
    (2015) Study on the anticarcinogenic efficacy of withaferin-A in DEN induced hepatocellular carcinoma: morphology and histopathology. Int J Health Sci Res 5:273–282.
    OpenUrl
  42. ↵
    1. Noori MS,
    2. Courreges MC,
    3. Bergmeier SC,
    4. McCall KD, and
    5. Goetz DJ
    (2020) Modulation of LPS-induced inflammatory cytokine production by a novel glycogen synthase kinase-3 inhibitor. Eur J Pharmacol 883:173340.
    OpenUrl
  43. ↵
    1. Oh JH,
    2. Lee TJ,
    3. Park JW, and
    4. Kwon TK
    (2008) Withaferin A inhibits iNOS expression and nitric oxide production by Akt inactivation and down-regulating LPS-induced activity of NF-kappaB in RAW 264.7 cells. Eur J Pharmacol 599:11–17.
    OpenUrlCrossRefPubMed
  44. ↵
    1. Palliyaguru DL,
    2. Chartoumpekis DV,
    3. Wakabayashi N,
    4. Skoko JJ,
    5. Yagishita Y,
    6. Singh SV, and
    7. Kensler TW
    (2016) Withaferin A induces Nrf2-dependent protection against liver injury: role of Keap1-independent mechanisms. Free Radic Biol Med 101:116–128.
    OpenUrl
  45. ↵
    1. Patel DP,
    2. Yan T,
    3. Kim D,
    4. Dias HB,
    5. Krausz KW,
    6. Kimura S, and
    7. Gonzalez FJ
    (2019) Withaferin A improves nonalcoholic steatohepatitis in mice. J Pharmacol Exp Ther 371:360–374.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Patel SB,
    2. Rao NJ, and
    3. Hingorani LL
    (2016) Safety assessment of Withania somnifera extract standardized for withaferin A: acute and sub-acute toxicity study. J Ayurveda Integr Med 7:30–37.
    OpenUrlCrossRef
  47. ↵
    1. Patil D,
    2. Gautam M,
    3. Mishra S,
    4. Karupothula S,
    5. Gairola S,
    6. Jadhav S,
    7. Pawar S, and
    8. Patwardhan B
    (2013) Determination of withaferin A and withanolide A in mice plasma using high-performance liquid chromatography-tandem mass spectrometry: application to pharmacokinetics after oral administration of Withania somnifera aqueous extract. J Pharm Biomed Anal 80:203–212.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Pires N,
    2. Gota V,
    3. Gulia A,
    4. Hingorani L,
    5. Agarwal M, and
    6. Puri A
    (2020) Safety and pharmacokinetics of withaferin-A in advanced stage high grade osteosarcoma: a phase I trial. J Ayurveda Integr Med 11:68–72.
    OpenUrlCrossRef
  49. ↵
    1. Purushotham PM,
    2. Kim JM,
    3. Jo EK, and
    4. Senthil K
    (2017) Withanolides against TLR4-activated innate inflammatory signalling pathways: a comparative computational and experimental study. Phytother Res 31:152–163.
    OpenUrl
  50. ↵
    1. Rosazza JP,
    2. Nicholas AW, and
    3. Gustafson ME
    (1978) Microbial transformations of natural antitumor agents. 7. 14-alpha-hydroxylation of withaferin-A by cunninghamella elegans (NRRL 1393). Steroids 31:671–679.
    OpenUrlPubMed
  51. ↵
    1. Saleem S,
    2. Muhammad G,
    3. Hussain MA,
    4. Altaf M, and
    5. Bukhari SNA
    (2020) Withania somnifera L.: insights into the phytochemical profile, therapeutic potential, clinical trials, and future prospective. Iran J Basic Med Sci 23:1501–1526.
    OpenUrl
  52. ↵
    1. Savai J,
    2. Varghese A, and
    3. Pandita N
    (2013) Lack of the cytochrome P450 3A interaction of methanolic extract of Withania somnifera, withaferin A, withanolide A and withanoside IV. J Pharm Negat Results 4:26–32.
    OpenUrl
  53. ↵
    1. Savai J,
    2. Varghese A,
    3. Pandita N, and
    4. Chintamaneni M
    (2015) In vitro assessment of CYP1A2 and 2C9 inhibition potential of Withania somnifera and Centella asiatica in human liver microsomes. Drug Metab Pers Ther 30:137–141.
    OpenUrl
  54. ↵
    1. Sayed N,
    2. Khurana A,
    3. Saifi MA,
    4. Singh M, and
    5. Godugu C
    (2019) Withaferin A reverses bile duct ligation-induced liver fibrosis by modulating extracellular matrix deposition: role of LOXL2/Snail1, vimentin, and NFκB signaling. Biofactors 45:959–974.
    OpenUrl
  55. ↵
    1. Schwabe RF,
    2. Tabas I, and
    3. Pajvani UB
    (2020) Mechanisms of fibrosis development in nonalcoholic steatohepatitis. Gastroenterology 158:1913–1928.
    OpenUrlPubMed
  56. ↵
    1. Sheka AC,
    2. Adeyi O,
    3. Thompson J,
    4. Hameed B,
    5. Crawford PA, and
    6. Ikramuddin S
    (2020) Nonalcoholic steatohepatitis: a review. JAMA 323:1175–1183.
    OpenUrl
  57. ↵
    1. Shiragannavar VD,
    2. Gowda NGS,
    3. Kumar DP,
    4. Mirshahi F, and
    5. Santhekadur PK
    (2021) Withaferin A acts as a novel regulator of liver X receptor-α in HCC. Front Oncol 10:628506.
    OpenUrl
  58. ↵
    1. Shohat B,
    2. Gitter S,
    3. Abraham A, and
    4. Lavie D
    (1967) Antitumor activity of withaferin A (NSC-101088). Cancer Chemother Rep 51:271–276.
    OpenUrl
  59. ↵
    1. Shohat B,
    2. Gitter S, and
    3. Lavie D
    (1970) Effect of withaferin A on Ehrlich ascites tumor cells-cytological observations. Int J Cancer 5:244–252.
    OpenUrlCrossRefPubMed
  60. ↵
    Siddharth S, Muniraj N, Saxena NK, and Sharma D (2019) Concomitant inhibition of cytoprotective autophagy augments the efficacy of withaferin A in hepatocellular carcinoma. Cancers (Basel) 11:453.
    OpenUrl
  61. ↵
    1. Sun GY,
    2. Li R,
    3. Cui J,
    4. Hannink M,
    5. Gu Z,
    6. Fritsche KL,
    7. Lubahn DB, and
    8. Simonyi A
    (2016) Withania somnifera and its withanolides attenuate oxidative and inflammatory responses and up-regulate antioxidant responses in BV-2 microglial cells. Neuromolecular Med 18:241–252.
    OpenUrl
  62. ↵
    1. Sun R,
    2. Kong B,
    3. Yang N,
    4. Cao B,
    5. Feng D,
    6. Yu X,
    7. Ge C,
    8. Feng S,
    9. Fei F,
    10. Huang J, et al.
    (2021) The hypoglycemic effect of berberine and berberrubine involves modulation of intestinal farnesoid x receptor signaling pathway and inhibition of hepatic gluconeogenesis. Drug Metab Dispos 49:276–286.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. Sun R,
    2. Yang N,
    3. Kong B,
    4. Cao B,
    5. Feng D,
    6. Yu X,
    7. Ge C,
    8. Huang J,
    9. Shen J,
    10. Wang P, et al.
    (2017) Orally administered berberine modulates hepatic lipid metabolism by altering microbial bile acid metabolism and the intestinal FXR signaling pathway. Mol Pharmacol 91:110–122.
    OpenUrlAbstract/FREE Full Text
  64. ↵
    1. Sun S and
    2. Wesolowski SS
    (2021) Biologically active metabolites in drug discovery. Bioorg Med Chem Lett 48:128255.
    OpenUrl
  65. ↵
    1. Surh YJ
    (2011) Reverse pharmacology applicable for botanical drug development—inspiration from the legacy of traditional wisdom. J Tradit Complement Med 1:5–7.
    OpenUrl
  66. ↵
    1. Tekula S,
    2. Khurana A,
    3. Anchi P, and
    4. Godugu C
    (2018) Withaferin-A attenuates multiple low doses of streptozotocin (MLD-STZ) induced type 1 diabetes. Biomed Pharmacother 106:1428–1440.
    OpenUrl
  67. ↵
    1. Thaiparambil JT,
    2. Bender L,
    3. Ganesh T,
    4. Kline E,
    5. Patel P,
    6. Liu Y,
    7. Tighiouart M,
    8. Vertino PM,
    9. Harvey RD,
    10. Garcia A, et al.
    (2011) Withaferin A inhibits breast cancer invasion and metastasis at sub-cytotoxic doses by inducing vimentin disassembly and serine 56 phosphorylation. Int J Cancer 129:2744–2755.
    OpenUrlCrossRefPubMed
  68. ↵
    1. Vanden Berghe W,
    2. Sabbe L,
    3. Kaileh M,
    4. Haegeman G, and
    5. Heyninck K
    (2012) Molecular insight in the multifunctional activities of withaferin A. Biochem Pharmacol 84:1282–1291.
    OpenUrlCrossRefPubMed
  69. ↵
    1. Varghese A,
    2. Pandita N, and
    3. Gaud RS
    (2014) In vitro and in vivo evaluation of CYP1A interaction potential of terminalia arjuna bark. Indian J Pharm Sci 76:138–147.
    OpenUrl
  70. ↵
    1. Vedi M,
    2. Rasool M, and
    3. Sabina EP
    (2014) Amelioration of bromobenzene hepatotoxicity by Withania somnifera pretreatment: role of mitochondrial oxidative stress. Toxicol Rep 1:629–638.
    OpenUrl
  71. ↵
    1. Vedi M and
    2. Sabina EP
    (2016) Assessment of hepatoprotective and nephroprotective potential of withaferin A on bromobenzene-induced injury in Swiss albino mice: possible involvement of mitochondrial dysfunction and inflammation. Cell Biol Toxicol 32:373–390.
    OpenUrl
  72. ↵
    1. Wang L,
    2. Zhu L,
    3. Gao S,
    4. Bao F,
    5. Wang Y,
    6. Chen Y,
    7. Li H, and
    8. Chen L
    (2018) Withanolides isolated from Nicandra physaloides protect liver cells against oxidative stress-induced damage. J Funct Foods 40:93–101.
    OpenUrl
  73. ↵
    1. Wang YX,
    2. Ding WB, and
    3. Dong CW
    (2015) Withaferin A suppresses liver tumor growth in a nude mouse model by downregulation of cell signaling pathway leading to invasion and angiogenesis. Trop J Pharm Res 14:1005-1011.
    OpenUrl
  74. ↵
    1. Wu J,
    2. Li X,
    3. Zhao J,
    4. Wang R,
    5. Xia Z,
    6. Li X,
    7. Liu Y,
    8. Xu Q,
    9. Khan IA, and
    10. Yang S
    (2018) Anti-inflammatory and cytotoxic withanolides from Physalis minima. Phytochemistry 155:164–170.
    OpenUrl
  75. ↵
    1. Wu KC and
    2. Lin CJ
    (2019) The regulation of drug-metabolizing enzymes and membrane transporters by inflammation: evidences in inflammatory diseases and age-related disorders. Yao Wu Shi Pin Fen Xi 27:48–59.
    OpenUrl
  76. ↵
    1. Xia Y,
    2. Wang P,
    3. Yan N,
    4. Gonzalez FJ, and
    5. Yan T
    (2021) Withaferin A alleviates fulminant hepatitis by targeting macrophage and NLRP3. Cell Death Dis 12:174.
    OpenUrl
  77. ↵
    1. Yan T,
    2. Wang H,
    3. Cao L,
    4. Wang Q,
    5. Takahashi S,
    6. Yagai T,
    7. Li G,
    8. Krausz KW,
    9. Wang G,
    10. Gonzalez FJ, et al.
    (2018) Glycyrrhizin alleviates nonalcoholic steatohepatitis via modulating bile acids and meta-inflammation. Drug Metab Dispos 46:1310–1319.
    OpenUrlAbstract/FREE Full Text
  78. ↵
    1. Yan T,
    2. Wang H,
    3. Zhao M,
    4. Yagai T,
    5. Chai Y,
    6. Krausz KW,
    7. Xie C,
    8. Cheng X,
    9. Zhang J,
    10. Che Y, et al.
    (2016) Glycyrrhizin protects against acetaminophen-induced acute liver injury via alleviating tumor necrosis factor-α mediated apoptosis. Drug Metab Dispos 44:720–731.
    OpenUrlAbstract/FREE Full Text
  79. ↵
    1. Yan T,
    2. Yan N,
    3. Wang P,
    4. Xia Y,
    5. Hao H,
    6. Wang G, and
    7. Gonzalez FJ
    (2020) Herbal drug discovery for the treatment of nonalcoholic fatty liver disease. Acta Pharm Sin B 10:3–18.
    OpenUrl
  80. ↵
    1. Yoon MY,
    2. Kim SJ,
    3. Lee BH,
    4. Chung JH, and
    5. Kim YC
    (2006) Effects of dimethylsulfoxide on metabolism and toxicity of acetaminophen in mice. Biol Pharm Bull 29:1618–1624.
    OpenUrlCrossRefPubMed
  81. ↵
    1. Younes R and
    2. Bugianesi E
    (2019) NASH in lean individuals. Semin Liver Dis 39:86–95.
    OpenUrlCrossRefPubMed
  82. ↵
    1. Zaher H,
    2. Buters JT,
    3. Ward JM,
    4. Bruno MK,
    5. Lucas AM,
    6. Stern ST,
    7. Cohen SD, and
    8. Gonzalez FJ
    (1998) Protection against acetaminophen toxicity in CYP1A2 and CYP2E1 double-null mice. Toxicol Appl Pharmacol 152:193–199.
    OpenUrlCrossRefPubMed
  83. ↵
    1. Zhang Y,
    2. Lu Y,
    3. Ji H, and
    4. Li Y
    (2019) Anti-inflammatory, anti-oxidative stress and novel therapeutic targets for cholestatic liver injury. Biosci Trends 13:23–31.
    OpenUrl
PreviousNext
Back to top

In this issue

Drug Metabolism and Disposition: 50 (5)
Drug Metabolism and Disposition
Vol. 50, Issue 5
1 May 2022
  • Table of Contents
  • About the Cover
  • Index by author
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Drug Metabolism & Disposition article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Withaferin A in the Treatment of Liver Diseases: Progress and Pharmacokinetic Insights
(Your Name) has forwarded a page to you from Drug Metabolism & Disposition
(Your Name) thought you would be interested in this article in Drug Metabolism & Disposition.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Review ArticleSpecial Section on Drug Metabolism in Liver Injury and Repair

Hepatoprotective Effects and Metabolism of Withaferin A

Yangliu Xia, Mingrui Yan, Ping Wang, Keisuke Hamada, Nana Yan, Haiping Hao, Frank J. Gonzalez and Tingting Yan
Drug Metabolism and Disposition May 1, 2022, 50 (5) 685-693; DOI: https://doi.org/10.1124/dmd.121.000455

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Review ArticleSpecial Section on Drug Metabolism in Liver Injury and Repair

Hepatoprotective Effects and Metabolism of Withaferin A

Yangliu Xia, Mingrui Yan, Ping Wang, Keisuke Hamada, Nana Yan, Haiping Hao, Frank J. Gonzalez and Tingting Yan
Drug Metabolism and Disposition May 1, 2022, 50 (5) 685-693; DOI: https://doi.org/10.1124/dmd.121.000455
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Natural Products in W. Somnifera
    • Conclusion
    • Acknowledgments
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Editorial: Drug Metabolism in Liver Injury and Repair
  • Novel Clinical DILI Biomarkers
Show more Special Section on Drug Metabolism in Liver Injury and Repair

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About DMD
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Journal of Pharmacology and Experimental Therapeutics
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-009X (Online)

Copyright © 2022 by the American Society for Pharmacology and Experimental Therapeutics