Next Article in Journal
Erythromycin Restores Osteoblast Differentiation and Osteogenesis Suppressed by Porphyromonas gingivalis Lipopolysaccharide
Next Article in Special Issue
Valorization of Punica granatum L. Leaves Extracts as a Source of Bioactive Molecules
Previous Article in Journal
Hesperidin Mitigates Cyclophosphamide-Induced Testicular Dysfunction via Altering the Hypothalamic Pituitary Gonadal Axis and Testicular Steroidogenesis, Inflammation, and Apoptosis in Male Rats
Previous Article in Special Issue
Chemical Constituents, Anticancer and Anti-Proliferative Potential of Limonium Species: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Autophagy Induction by Scutellaria Flavones in Cancer: Recent Advances

1
Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (University), Mullana, Ambala 133207, India
2
Department of Botany, Central University of Jammu, Samba 181143, India
3
Punjab Biotechnology Incubator (P.B.T.I.), Phase VIII, Mohali 160071, India
4
The Basic Research Laboratory, Center for Cancer Research, National Institutes of Health, Frederick, MD 20892, USA
5
NGO Praeventio, 50407 Tartu, Estonia
6
University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali 140413, India
7
Advance Cardiac Centre Department of Cardiology, Post Graduate Institute of Medical Education and Rsearch (P.G.I.M.E.R.), Chandigarh 160012, India
8
Department of Pharmacology, School of Health Sciences & Technology (SoHST), University of Petroleum and Energy Studies, Bidholi, Dehradun 248007, India
9
Department of Research, Sir Ganga Ram Hospital, New Delhi 122016, India
10
Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
11
Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut P.O. Box 13-5053, Lebanon
12
Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
13
Nutrition and Bromatology Group, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E32004 Ourense, Spain
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(2), 302; https://doi.org/10.3390/ph16020302
Submission received: 16 December 2022 / Revised: 21 January 2023 / Accepted: 8 February 2023 / Published: 15 February 2023
(This article belongs to the Special Issue Anticancer Compounds in Medicinal Plants 2023)

Abstract

:
In parallel with a steady rise in cancer incidence worldwide, the scientific community is increasingly focused on finding novel, safer and more efficient modalities for managing this disease. Over the past decades, natural products have been described as a significant source of new structural leads for novel drug candidates. Scutellaria root is one of the most studied natural products because of its anticancer potential. Besides just describing the cytotoxic properties of plant constituents, their molecular mechanisms of action in different cancer types are equally important. Therefore, this review article focuses on the role of the Scutellaria flavones wogonin, baicalein, baicalin, scutellarein and scutellarin in regulating the autophagic machinery in diverse cancer models, highlighting these molecules as potential lead compounds for the fight against malignant neoplasms. The knowledge that autophagy can function as a dual-edged sword, acting in both a pro- and antitumorigenic manner, further complicates the issue, revealing an amazing property of flavonoids that behave either as anti- or proautophagic agents.

1. Introduction

In ethnomedicine, plants have been used for the treatment of various health problems for centuries [1]. Even today, more than 80% of the world’s population relies on herbal medicines for their primary health care [2]. Considering malignant disorders, a more systematic approach to herbal medicines started with the launch of a large-scale screening program by the U.S. National Cancer Institute in 1960, leading to the isolation and characterization of several novel anticancer agents and inspiring researchers all over the world to investigate the infinite molecular abundance of the plant kingdom [3,4,5].
Scutellaria, a genus of flowering plants from the family Lamiaceae that is usually known as skullcaps, is one of the most attractive herbal resources of polyphenolic compounds with anticancer properties. Indeed, several important phytochemicals from the class of flavonoids have been isolated from the roots of Scutellariae plants, including wogonin, wogonoside, baicalein, baicalin, scutellarein and scutellarin (Figure 1) [6,7]. The various anticancer mechanisms of these flavones have been described in different cancer cell cultures and xenografted animal preclinical models, demonstrating the potential of these compounds to exert anti-inflammatory, cell death-inducing, antimetastatic and antiangiogenic properties in different cancer types [8,9,10,11]. Furthermore, among several other mechanisms, Scutellaria flavones have been shown to suppress malignant growth through the modulation of autophagy [12,13]. Moreover, depending on various factors, such as the cancer cell type, activated cellular signaling network and molecular characteristics of the isolated compounds, Scutellaria-derived flavonoids can exert antitumor activity [13,14] by inducing autophagic processes [12,15]. The present review focused on the potential of Scutellaria flavones to regulate the autophagic machinery in diverse cancer models. Flavones are the major group of naturally occurring phytochemical substances that have attracted substantial attention due to their chemotherapeutic and/or pharmacological properties [16]. Scutellaria flavones are generally derived from the Scutellaria baicalensis Georgi that show many pharmacological properties, such as antioxidation, antiviral, hepatoprotective, etc. [17], and Scutellaria flavones have shown apoptosis-inducing properties in many cancer cell lines [18].
Numerous aspects of Scutellaria flavones related to autophagy death in cancer cells are covered in this review. The authors surveyed the literature, and few reviews showed the autophagy-targeting pathways in the cancer models using Scutellaria flavones. That is why the authors highlighted the autophagy-signaling pathways modulated by Scutellaria flavones.
In this review article, the chemical features of the flavone-type compounds of the Scutellaria root are described by analyzing their autophagy-modulating abilities in diverse malignant models to better understand the complex anticancer mechanisms of these agents in different malignant systems. In addition, the absorption and metabolic profiles of these important molecules within in vivo systems are discussed to present, in addition to attractive anticancer effects, the potential bottlenecks for initiating clinical trials in the future.

2. Sources and Chemistry of Scutellaria Flavones (Wogonin, Baicalein, Baicalin, Scutellarein and Scutellarin)

2.1. Wogonin

Plants possess a plethora of secondary metabolites, including polyphenolic flavonoids. They are naturally distributed in the pigments, flowers, stems, seeds, fruits, vegetables, spices and flowers [19]. Humans are exposed to these phytonutrients of therapeutic importance through various sources. Among the various flavonoids, 5, 7-dihydroxy-8-methoxy flavone, also known as wogonin, is an important derivative of flavones. In the year 1930, wogonin was first isolated from Scutellaria baicalensis (Lamiaceae), which is a widely distributed family in North America, Russia and East Asia [20]. Wogonin naturally occurs in the whole herb of S. baicalensis, including the roots [21], and in various plants, such as Andrographis paniculata Burm.f (leaves), Anodendron affine (Hook. & Arn.) Druce (stem) [22,23] and Tetracera indica L. (leaves and stem) [20]. It can be synthesized/obtained via extraction, a chemical process involving cyclization of 1,3- diaryl-diketone, or with Wessely–Moser rearrangement [24]. It has been observed that the chemical synthesis of this compound is a convenient method to obtain it in large quantities, which involves using trimethoxyphenol as the starting material [25].
Wogonin is a monoflavonoid with a molecular weight of 284.267 g/mol and solubility in organic solvents, such as ethanol, dimethylformamide (D.M.F.) and dimethyl sulfoxide (DMSO). Interestingly, many of the therapeutic properties of the Scutellaria species viz., including antioxidant (Gao et al., 1999), antiviral [26], anti-inflammatory [27], antiproliferative [6] and multi-drug resistant [28], can be attributed to wogonin along with other phytoconstituents (Table 1).

2.2. Baicalein

Baicalein (5,6,7-trihydroxyflavone) is one of the naturally occurring flavones, an important class of flavonoids, originally isolated and extracted from Scutellaria baicalensis and Scutellaria lateriflora [29,30]. It has also been reported in Oroxylum indicum and Thyme. Baicalein is a structural derivative of 1-benzopyran phenylpropanoid and a functional parent of “Bicalin” [31]. Chemically, it is an aglycone of “baicalin” having a hydroxyl group at the C-5, C-6 and C-7 positions. Its molecular structure shows the presence of di-orthohydroxyl as a functional group associated with the ring –A. Interestingly, this phytocompound is a key ingredient of Sho–Saiko To, an herbal formula of Chinese origin, and is also related to “Kempo medicine“ of Japanese origin. Baicalein acts as a “positive allosteric modulator” of the GABAA receptor at the benzodiazepine/non-benzodiazepine site (Table 1) [32]. It also exhibits selectivity for the α2 and α3 subunits in the GABAA receptors due to its structural feature, which is responsible for its anxiolytic property [29].

2.3. Baicalin

One of the most important phytoconstituents of Scutellaria baicalensis is baicalin, also known as 5,6-dihydroxy-4-oxygen-2-phenyl-4H-1-benzopyran-7-β-D-glucopyranose acid [33]. It is the critical constituent of the Scutellaria species and is crucial to its pharmacological and therapeutic functions [34]. “Baicalin” is present in abundant quantities in the stem and leaves of the Scutellaria species of the Lamiaceae family. Interestingly, Baicalin has been utilized for centuries as a traditional Chinese herbal medicine for curing psoriasis, inflammation, hypertension, cardiovascular diseases, etc. [35]. Other plant species in which it is present and isolated are S. lateriflora, S. galericulata and Thalictrum baicalense [36]. Baicalin has also been reported to have been purified from Radix scutellariae via the process of uridine diphosphate glucuronidation. Moreover, it is a “flavone glycoside” and, upon further hydrolysis, is converted to an aglycone “baicalein” [37,38]. It is also called glycosyloxyflavone, which represents the “7-O-glucuronide” of baicalein and the conjugated acid of a baicalin(1-). The various bioprotective roles of baicalin include its ability to act as a neuroprotective, cardioprotective, antioxidant, antiatherosclerotic, antibacterial and anticoronaviral agent [39] (Table 1).

2.4. Scutellarein

Scutellarein is a naturally occurring flavone, effectively isolated from Scutellaria baicalensis Georgi along with Scutellaria lateriflora, Scoparia dulcis, Artemisia douglasiana and Asplenium belangeri (Fern) [40]. It is chemically designated as 6-hydroxyapigenin or 4’,5,6,7-tetrahydroxyflavanone and shows the presence of hydroxy groups at the C-4’, -5, -6 and -7 positions. Moreover, it is functionally related to apigenin [41]. Recent research has focused on scutellarein owing to its interesting bioprotective properties, including anticancer, antiproliferative and antioxidant activities [42] (Table 1). Interestingly, being a flavone, its structure is based upon the backbone of -phenylchromen-4-one (2-phenyl-1-benzopyran-4-one) [43]. Therefore, it is known as a lipid molecule. The solubility of scutellarein is observed to be less in water and shows weak acidic properties. Furthermore, it is efficiently produced from “apigenin” [36]. Apigenin, 4’,5,7-trihydroxy-flavone, is a monomeric flavonoid found in the daily diet [44]. Apigenin has gained attention among researchers partly due to being nonmutagenic and of low toxicity compared to related flavonoids [45]. Scutellarein was used to prepare other compounds, such as scutellarin, 4’,6-dihydroxy-5,7-dimethoxyflavone and 6-hydroxy-4’,5,7-trimethoxyflavone.

2.5. Scutellarin

Scutellarin is a flavonoid frequently derived from the genus Scutellaria and Erigeron from Asteraceae [32]. It is widely used in the preparation of herbal medicine due to its numerous pharmacological properties [38] (Table 1). Chemically, it is a glucuronide conjugate of 5,6,7,4′-tetrahydroxyflavone at the “7-O position”. Scutellarin is a monosaccharide derivative whose chemical analogs share a common skeleton of the flavonoid/s. Scutellarin, a phenolic natural compound, is extensively used to treat various ailments, viz., liver diseases [46,47], cerebrovascular diseases [48] and hyperlipidemia [49,50]. Recent reports have indicated that over 10 million Chinese patients depend on scutellarin and associated drugs, indicating its therapeutic potency [51].

3. Absorption and Metabolism of Scutellaria Flavones

Since flavonoids are important phytoconstituents of plants, it is essential for their distribution, absorption and metabolism in plasma and tissue after ingestion. Therefore, this approach helps determine the concentrations and forms of such compounds in plasma and tissue. In addition, most flavonoids tend to bind sugars as b-glycosides, which further determine their absorption directly from the small intestine or through the colon [42]. Interestingly, before absorption, the dietary flavonoids must be released by chewing or digestion via digestive juices (gastrointestinal tract) followed by the action of microorganisms in the colon. Overall, it depends upon the physio-chemical properties of the flavonoids, including the molecular size, configuration, solubility, lipophilicity, etc.
After being absorbed by the small intestine, flavonoids are often conjugated with glucuronic acid. Due to the absence of free flavonoid aglycones in the plasma or urine, efficient absorption is achieved. When aglycones and glucosides are taken together, isoflavones are found to have the best bioavailability of all the subclasses of flavonoids. In contrast, anthocyanins have the lowest bioavailability and are quickly absorbed. Moreover, the elimination half-lives of different flavonoid subclasses vary from flavonols [53,54].
Importantly, the metabolism of flavonoids involves two key compartments: the small intestine, liver and kidneys, which represent the first compartment, and the colon, which constitutes the second compartment [55]. The flavonoids absorbed (followed by bile secretion) and unabsorbed (from the small intestine) reach the colon, which is a crucial step in the overall metabolism of these constituents. In the first section/compartment, the flavonoids and their colonic metabolites are biotransformed through enzymatic action, as evident from the conjugation of polar hydroxyl groups with glucuronic acid [53]. Additionally, the process of O-methylation plays a critical role in the inactivation of the catechol moiety if present. Additionally, recent research has revealed that glycosides are deglycosylated in the intestine [56]. As a result, after ingesting flavonoids, the conjugated metabolites of these compounds can be identified in the plasma. Most flavonoids prefer/undergo sulfation, methylation and glucuronidation in the small intestine and liver. Fascinatingly, flavonoids are found to have quick urine and biliary excretion but low intestinal absorption [57]. Keeping this in mind, the present review highlighted the various pharmacokinetic studies involving flavonoids from Scutellaria that are documented in a tabular form (Table 2).

4. Mechanistic Role of Autophagic Death in Cancer

According to the type of tumor and stage of oncogenic development, autophagy can –play a dual-edged sword role by acting as both pro- and antitumorigenic (sikder et al.; 2022 [66]). However, the role of autophagy in cancer is controversial and complicated.

4.1. Antitumorigenic

During the early stages of cancer, autophagy plays an important role in removing aggregated misfolded proteins, damaged mitochondria and other cellular organelles, thereby protecting the cells from further genomic instability [77,78,79]. In addition, autophagy may initiate the cell death machinery in the cancer cells with impaired apoptotic cell death. Growing solid tumors that have an autophagy defect at early stages is reported to show chemotherapeutic drug resistance. Therefore, the induction of autophagy may increase cell death either by mitophagy or by apoptosis. In vitro studies have shown that EB1089 (an analogue of vitamin D) and arsenic trioxide can promote autophagy-induced apoptotic cell death [80,81]. Treatment with specific chemotherapeutic agents, such as dexamethasone, etoposide and fenretinide, induces autophagy-mediated cell death in vitro [82,83,84]. These autophagy-inducing drugs act by blocking the activity of mTOR, possibly by activating AMPK. Rapamycin is reported to stabilize the raptor–mTOR complex by binding with FKBP12 and, thus, suppress mTOR activity, resulting in autophagy induction [85]. Similar findings are also reported in neuroblastoma cells, hepatocellular carcinoma (HCC) and murine sarcoma, where autophagy-mediated cell-cycle arrest suppressed tumor growth [86,87,88]. Antioncogenes connected to autophagy, such as p53 and Beclin-1, as well as phosphatase and the tensin homolog on chromosome 10 (PTEN) (Figure 2) play important roles in controlling carcinogenesis. For instance, the growth of autophagosomes depends on Beclin-1 [89,90]. PTEN initiates autophagy and inhibits the PI3K/Akt/mTOR pathway, controlling the cell cycle and cell proliferation [90,91]. By activating TSC1/2 and AMP-activated protein kinase, p53 regulates autophagy and inhibits mTOR [90].

4.2. Protumorigenic

Autophagy helps tumor cells survive by providing higher amounts of nutrients and oxygen during metabolic and hypoxic stress [92,93,94]. Moreover, Beclin1-UV irradiation-resistance-associated gene (UVRAG) core complex-induced autophagy confers resistance to radiation-induced DNA double-strand breaks (DSBs) in tumor cells [95,96]. Paglin et al. first observed the formation of acidic vacuoles in neoplastic cells when exposed to IR that helps tumor cells survive [95]. Interestingly, the role of autophagy in tumor cell survival is not only restricted to the tumor vicinity, while hypoxia inducible factor-1α (HIF-1α) induced autophagy also plays an important role in metastasis [97]. Autophagy can induce resistance to various cancer therapies, leading to chemo resistance and cancer cell survival [98,99,100]. Therefore, inhibition of autophagy with various pharmacological inhibitors, such as 3-methyladenine (3-MA, a PI3K III inhibitor), Bafilomycin A (inhibitor of vacuolar-type H+-ATPase), chloroquine (CQ) or hydroxychloroquine (HCQ) (impair autophagosome fusion with lysosomes), or with genetic deletion of autophagy-related genes, such as ATG5, ATG6 and ATG7, enhance the effect of various anticancer therapies [101,102,103,104,105].
Numerous studies demonstrated that the activation of tyrosine kinases (T.K.s) and receptor tyrosine kinases (RTKs) that include EGFR, PDGFR, RAF and VEGFR modifies autophagy by triggering various signaling pathways, such as PI3K/AKT/mTORC1 and RAS/MAPK [106,107]. While autophagy functions in the wake of RTK pathways, there is also a reciprocal relationship between autophagy and RTK signaling. RTKs can restrict autophagy through mTORC1, while autophagy can also favorably influence RTK signaling through activation of mTORC2 [108].
Effective antitumor therapies have been created using tyrosine kinase inhibitors (T.K.I.s). The catalytic domain of tyrosine kinase is where T.K.I.s and ATP fight for a binding site (T.K.s) [109]. The F.D.A. has authorized numerous T.K.I. drugs, including imatinib (PDGFR, ABL kinase), mesylate, erlotinib and gefitinib (EGFR TKIs), lapatinib (EGFR/HER2), Sunitinib (VEGF, PDGFR) and sorafenib (VEGFR kinase, RAF, PDGFR), for use in human clinical trials [110,111].
As autophagy has both pro- and anticancer effects, proper tumor stage and tumor type need to be considered accurately to answer the question whether to induce or suppress autophagy. Many clinical trials targeting autophagy are now being conducted to increase the effectiveness of medicines that modulate autophagy to treat cancer.

5. Regulation of Autophagy by Scutellaria Flavones

5.1. Wogonin and Autophagy Induction

Wogonin is a flavonoid obtained from Scutellaria baicalensis; studies have exhibited the prominent role of wogonin in inhibiting malignant tumor growth via inducing apoptosis and regulating autophagy [112,113]. Autophagy plays an important role in disassembling the unnecessary or dysfunctional components of the cell. It acts as a regulatory and destructive mechanism [12,114,115]. Studies show that autophagy contributes to tumor suppression via different inhibiting mechanisms [114,115,116]. Studies show the role of wogonin in strengthening the efficacy of anticancer drugs and offering lower toxicity when used in combination. Different studies highlight the role of Unc-51-like autophagy-activating kinase 1 (ULK1), protein kinase B (AKT), eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), mammalian target of rapamycin (mTOR) and cylindromatosis (CYLD) in mediating autophagy. Investigations demonstrate the role of wogonin in upregulating and downregulating the expression of these molecules involved in autophagy in human pancreatic cancer cells (HPCCs) [116,117,118,119]. It was shown that wogonin regulated autophagy via ULK1, as it is the key regulator of autophagy. It coordinates the earliest phases of autophagosome production when combined with oxaliplatin to stimulate autophagy in human gastric (BGC-823) cells. The study showed that wogonin reduced the phosphorylation necessary for mitochondrial homeostasis and cell survival during starvation, either alone or in combination with oxaliplatin. The study’s findings showed that combining oxaliplatin and wogonin promotes excessive autophagy by excessive nitrosative stress, amplifying oxaliplatin-induced cell death. Studies report that wogonin promotes reactive oxygen species (ROS) formation in cancer cells, and the overgeneration of ROS in human gastric cancer cells is linked with autophagy [120]. Zhang et al. reported that wogonin and icotinib, when used in combination, activated autophagy and increased the rate of apoptosis in lung cancer cells (NCI-H1975), suggesting the role of autophagy in apoptosis. The study demonstrated that the combined effect of wogonin and icotinib increased the phosphorylation level of mTORC1, a classical regulator of autophagy, thereby promoting autophagy [121]. Li et al. also reported that wogonin significantly downregulated mTOR and upregulated ULK1, AKT, 4E-BP1 and CYLD expressions in human pancreatic cancer cells (HPCCs), throwing light on the role of wogonin-induced autophagy activators [119].
Further, the study demonstrated that wogonin activated Beclin-1/PI3K, enhancing ROS generation and ROS-mediated autophagy [119]. According to studies, wogonin controls the Bax/Bcl-2 and c-myc signaling pathway to cause apoptosis [122]. Many health benefits, including anticancer properties, are attributed to wogonin. Many in vitro studies emphasize the role of wogonin in the regulation of autophagy in cancer, and translating the beneficial properties of wogonin from bench side to bedside in cancer management could prove to be a useful strategy to exploit the therapeutic potential of wogonin (Table 2).

5.2. Baicalein and Baicalin Induce Autophagy

Scutellaria root contains the flavonoid derivative chemicals baicalein and baicalin. Both have potent antioxidant and anti-inflammatory activities and are often nontoxic to human cells [123,124]. Moreover, many studies have indicated their antitumor and antimetastatic roles in various cancer types [125,126,127]. Different mitochondrial/endoplasmic reticulum pathways mediated by reactive oxygen species and inhibition of AKT are responsible (Table 2) for the transmission of these effects [128,129,130,131]. For instance, Aryal et al. confirmed that baicalein induced autophagy in cancer cells rather than apoptosis. They showed that the baicalein-induced cell death was completely reversed when autophagy was suppressed by inhibiting the expression levels of molecules, such as Beclin-1, vacuolar protein sorting 34 (Vps34), autophagy-related (Atg)5 and Atg7, but not by a caspase inhibitor. Their findings suggested that baicalein enhanced the autophagic flow in addition to autophagosome formation. Moreover, their data suggested that the anticancer autophagic role of baicalein is mediated through upregulation of the AMPK/ULK1 pathway and downregulation of mTOR/Raptor complex 1 expression [132]. A similar study in ovarian cancer cells confirmed the role of baicalein in inducing Beclin-1-mediated autophagy. They also showed that baicalein-mediated autophagy is associated with extracellular signal-regulated kinase (ERK, Thr202/Thr204) and AKT (Ser473) phosphorylation [133]. Wang Z et al. reported that only baicalein, among four different flavonoids, showed an antitumor effect on hepatic cellular carcinoma (H.C.C.). They further showed the role of E.R. stress in inducing apoptosis and protective autophagy triggered by baicalein [134]. In a Lewis lung cancer (L.L.C.) xenograft model, a recent study demonstrated the important function of the A.M.P.K./mitochondrial fission pathway in regulating baicalein-induced apoptosis and autophagy. Even mitochondrial abnormalities were restored by reducing A.M.P.K. activation [135]. Together, these two studies indicate that the mitochondrial/E.R. axis plays a potent role in the baicalein-induced antitumor effect. Another intriguing study discovered that baicalein plays a dose- and time-dependent effect in triggering autophagy in undifferentiated follicular thyroid cancer cells (F.R.O.). Beclin-1, Atg5, p62 and Atg12 expression levels were significantly upregulated, and the ERK and PI3K/Akt pathways were inhibited. [136]. Baicalein was even shown to induce cell cycle arrest in cervical cancer cells by reducing the Cyclin D1 protein concentration and modulating the AKT/mTOR signaling pathway [137]. Aberrant activation of AKT serine/threonine protein kinase modulates autophagy in gastric cancer cells [138]. The synergistic role of baicalein along with cisplatin (DDP) has been shown to have promising antitumor effects on even DDP-resistant SGC-7901/DDP gastric cancer. The combined impact was more potent than DDP or baicalein acting separately. By triggering apoptosis and autophagy via the Akt/mTOR and Nrf2/Keap 1 pathways, baicalein increased the DDP sensitivity of SGC-7901/DDP gastric cancer cells [139]. In addition, baicalin also upregulated intercellular Ca2+ and ROS, associated with downregulating the PI3K/Akt/mTOR, ERK1/2 and β-catenin signaling pathways. Chelation of free Ca2+ with BAPTA-AM also downregulated apoptosis induction and ROS accumulation. Consequently, baicalin may be observed as a viable contender for the diagnosis of osteosarcoma [140].

5.3. Scutellarein and Scutellarin Induce Autophagy

Scutellarin and scutellarein are the natural oxyflavonoids obtained from Erigeron breviscapus; they are naturally glycosylated, perform a wide range of functions and have a variety of medicinal properties [13,141,142]. The therapeutic properties are as follows: vasodilating, acting as an anticoagulant and being tumor suppressive. Different cumulative reports show the repressive effects of scutellarin and scutellarein on cancers, such as liver, lung, colon, lymphoma, etc. Studies report that cutellarin and scutellarein induce apoptosis and trigger autophagy (Table 3) in cancer cells [52,143,144,145]. In a study conducted by Sun et al., scutellarin stimulated the phosphorylation of ERK1/2. Once phosphorylated and activated, it induced autophagy and apoptosis in PC-9 and H1975 cells [146]. Different studies show that combining scutellarin and scutellarein with commonly available anticancer drugs such as cisplatin enhances the sensitivity and efficacy of the commonly available anticancer compounds; in another study conducted by Sun et al., cisplatin, when combined with scutellarin, drastically inhibited the growth of cancer cells. Further, the study reported that scutellarin resulted in the activation of the p53 pathway mediated by ERK, which further resulted in caspase-3-mediated apoptosis. Further, the study reported that cisplatin-induced cytotoxic autophagy was enhanced by scutellarin [49,147,148,149,150]. Hou et al. demonstrated the role of scutellarin on the breast cancer cells MCF-7. The study demonstrated significant growth reduction in breast cancer cells treated with scutellarin. Additionally, it was found that scutellarin promoted apoptosis and autophagy by activating the HIPPO–YAP signaling pathway, supporting the potential therapeutic application of scutellarin-based drugs to provide patients with breast cancer the best possible outcomes [151,152]. The potential benefits of scutellarin and scutellarein need more clinical investigation for cancer treatment and as a newer therapeutic option [153,154,155,156,157].

6. Conclusions

In this review article, the molecular mechanisms of the flavone aglycones baicalein, scutellarein and wogonin and their glycosides baicalin and scutellarin derived from Scutellaria roots on the regulation of autophagy are discussed. Autophagy is known to play a dual-edged sword role in the carcinogenesis process, acting either as a protumorigenic or antitumorigenic mechanism, dependent on cancerous cell types and their degree of malignancy. Therefore, the studied flavones can behave as antiautophagic or proautophagic agents in suppressing tumorigenesis and malignant progression in diverse tumors. Therefore, understanding these fine cellular regulatory processes is important to better intervene in these mechanisms in the future, ultimately leading to safer and more efficient treatment modalities in the fight against different cancerous diseases.

Author Contributions

H.S.T., S.B., A.K., P.A., K.S., S.R., K.V., T.B., R.R. and K.S. contributed towards preparation of the original manuscript and during the peer-review process. H.S.T., S.H. and M.A.P. supervised the project. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Authors would like to thank BioRender software for drawing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Petrovska, B.B. Historical Review of Medicinal Plants’ Usage. Pharmacogn. Rev. 2012, 6, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ekor, M. The Growing Use of Herbal Medicines: Issues Relating to Adverse Reactions and Challenges in Monitoring Safety. Front. Pharmacol. 2014, 4, 177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sak, K. Anticancer Action of Plant Products: Changing Stereotyped Attitudes. Explor. Target Antitumor. Ther. 2022, 3, 423–427. [Google Scholar] [CrossRef]
  4. Hsieh, Y.-S.; Yang, S.-F.; Sethi, G.; Hu, D.-N. Natural Bioactives in Cancer Treatment and Prevention. Biomed. Res. Int. 2015, 2015, 182835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Salehi, B.; Quispe, C.; Imran, M.; Ul-Haq, I.; Živković, J.; Abu-Reidah, I.M.; Sen, S.; Taheri, Y.; Acharya, K.; Azadi, H.; et al. Nigella Plants—Traditional Uses, Bioactive Phytoconstituents, Preclinical and Clinical Studies. Front. Pharmacol. 2021, 12, 625386. [Google Scholar] [CrossRef] [PubMed]
  6. Li-Weber, M. New Therapeutic Aspects of Flavones: The Anticancer Properties of Scutellaria and Its Main Active Constituents Wogonin, Baicalein and Baicalin. Cancer Treat. Rev. 2009, 35, 57–68. [Google Scholar] [CrossRef]
  7. Shah, M.; Mubin, S.; Hassan, S.S.U.; Tagde, P.; Ullah, O.; Rahman, M.H.; Al-Harrasi, A.; Rehman, N.U.; Murad, W. Phytochemical Profiling and Bio-Potentiality of Genus Scutellaria: Biomedical Approach. Biomolecules 2022, 12, 936. [Google Scholar] [CrossRef] [PubMed]
  8. EghbaliFeriz, S.; Taleghani, A.; Tayarani-Najaran, Z. Scutellaria: Debates on the Anticancer Property. Biomed. Pharmacother. 2018, 105, 1299–1310. [Google Scholar] [CrossRef]
  9. Singh, S.; Meena, A.; Luqman, S. Baicalin Mediated Regulation of Key Signaling Pathways in Cancer. Pharmacol. Res. 2021, 164, 105387. [Google Scholar] [CrossRef]
  10. Liu, H.; Dong, Y.; Gao, Y.; Du, Z.; Wang, Y.; Cheng, P.; Chen, A.; Huang, H. The Fascinating Effects of Baicalein on Cancer: A Review. Int. J. Mol. Sci. 2016, 17, 1681. [Google Scholar] [CrossRef]
  11. Banik, K.; Khatoon, E.; Harsha, C.; Rana, V.; Parama, D.; Thakur, K.K.; Bishayee, A.; Kunnumakkara, A.B. Wogonin and Its Analogs for the Prevention and Treatment of Cancer: A Systematic Review. Phytother. Res. 2022, 36, 1854–1883. [Google Scholar] [CrossRef] [PubMed]
  12. Huynh, D.L.; Sharma, N.; Kumar Singh, A.; Singh Sodhi, S.; Zhang, J.J.; Mongre, R.K.; Ghosh, M.; Kim, N.; Ho Park, Y.; Kee Jeong, D. Anti-tumor activity of wogonin, an extract from Scutellaria baicalensis, through regulating different signaling pathways. Chin. J. Nat. Med. 2017, 15, 15–40. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, L.; Liu, T.; Tao, W.; Liao, N.; Yan, Q.; Li, L.; Tan, J.; Shen, W.; Cheng, H.; Sun, D. Flavonoids from Scutellaria Barbata D. Don Exert Antitumor Activity in Colorectal Cancer through Inhibited Autophagy and Promoted Apoptosis via ATF4/Sestrin2 Pathway. Phytomedicine 2022, 99, 154007. [Google Scholar] [CrossRef] [PubMed]
  14. Phan, T.; Nguyen, V.H.; A’lincourt Salazar, M.; Wong, P.; Diamond, D.J.; Yim, J.H.; Melstrom, L.G. Inhibition of Autophagy Amplifies Baicalein-Induced Apoptosis in Human Colorectal Cancer. Mol. Ther. Oncolytics 2020, 19, 1–7. [Google Scholar] [CrossRef]
  15. Kim, H.I.; Hong, S.H.; Ku, J.M.; Lim, Y.S.; Lee, S.J.; Song, J.; Kim, T.Y.; Cheon, C.; Ko, S.-G. Scutellaria Radix Promotes Apoptosis in Non-Small Cell Lung Cancer Cells via Induction of AMPK-Dependent Autophagy. Am. J. Chin. Med. 2019, 47, 691–705. [Google Scholar] [CrossRef]
  16. Bisol, Â.; Campos, P.S.; Lamers, M.L. Flavonoids as Anticancer Therapies: A Systematic Review of Clinical Trials. Phytother. Res. 2020, 34, 568–582. [Google Scholar] [CrossRef]
  17. Elkin, Y.N.; Kulesh, N.I.; Stepanova, A.Y.; Solovieva, A.I.; Kargin, V.M.; Manyakhin, A.Y. Methylated Flavones of the Hairy Root Culture Scutellaria Baicalensis. J. Plant Physiol. 2018, 231, 277–280. [Google Scholar] [CrossRef] [PubMed]
  18. Liao, H.; Ye, J.; Gao, L.; Liu, Y. The Main Bioactive Compounds of Scutellaria Baicalensis Georgi. for Alleviation of Inflammatory Cytokines: A Comprehensive Review. Biomed. Pharmacother. 2021, 133, 110917. [Google Scholar] [CrossRef]
  19. Timberlake, C.F.; Henry, B.S. Plant Pigments as Natural Food Colours. Endeavour 1986, 10, 31–36. [Google Scholar] [CrossRef]
  20. Shang, X.; He, X.; He, X.; Li, M.; Zhang, R.; Fan, P.; Zhang, Q.; Jia, Z. The Genus Scutellaria an Ethnopharmacological and Phytochemical Review. J. Ethnopharmacol. 2010, 128, 279–313. [Google Scholar] [CrossRef]
  21. Hui, K.M.; Huen, M.S.Y.; Wang, H.Y.; Zheng, H.; Sigel, E.; Baur, R.; Ren, H.; Li, Z.W.; Wong, J.T.-F.; Xue, H. Anxiolytic Effect of Wogonin, a Benzodiazepine Receptor Ligand Isolated from Scutellaria Baicalensis Georgi. Biochem. Pharmacol. 2002, 64, 1415–1424. [Google Scholar] [CrossRef] [PubMed]
  22. INAGAKI, I.; HISADA, S.; SHIMA, K. Studies on the Constituents of Anodendron Affine DURCE. I.: Isolation of Wogonin, Dambonitol, Sucrose and Some Other Components from Stems. Yakugaku Zasshi 1971, 91, 1133–1136. [Google Scholar] [CrossRef] [Green Version]
  23. Sukmawati, R.; Guntoro, D.; Junaedi, A. Flavone Content Analysis of Wogonin (5,7- Dihydroxy-8-Methoxy) from Tetracera Indica L. MERR. Rasayan J. Chem. 2020, 13, 1124–1128. [Google Scholar] [CrossRef]
  24. Wessely, F.; Moser, G.H. Synthese und Konstitution des Skutellareins. Monatsh. Chem. 1930, 56, 97–105. [Google Scholar] [CrossRef]
  25. Xia, Y.; Wu, C.; Liu, W.; Feng, F.; Li, Z. A Novel Process Related Impurity and Forced Degradation Study of Synthetic Wogonin: Development of a Liquid Chromatographic Method for Purity Control. J. Pharm. Biomed. Anal. 2012, 71, 168–172. [Google Scholar] [CrossRef] [PubMed]
  26. Gao, Z.; Huang, K.; Yang, X.; Xu, H. Free Radical Scavenging and Antioxidant Activities of Flavonoids Extracted from the Radix of Scutellaria Baicalensis Georgi. Biochim. Biophys. Acta Gen. Subj. 1999, 1472, 643–650. [Google Scholar] [CrossRef]
  27. Park, B.K.; Heo, M.Y.; Park, H.; Kim, H.P. Inhibition of TPA-Induced Cyclooxygenase-2 Expression and Skin Inflammation in Mice by Wogonin, a Plant Flavone from Scutellaria Radix. Eur. J. Pharmacol. 2001, 425, 153–157. [Google Scholar] [CrossRef] [PubMed]
  28. Lee, E.; Enomoto, R.; Koshiba, C.; Hirano, H. Inhibition of P-Glycoprotein by Wogonin Is Involved with the Potentiation of Etoposide-Induced Apoptosis in Cancer Cells. Ann. N. Y. Acad. Sci. 2009, 1171, 132–136. [Google Scholar] [CrossRef]
  29. Zhao, Z.; Nian, M.; Qiao, H.; Yang, X.; Wu, S.; Zheng, X. Review of Bioactivity and Structure-Activity Relationship on Baicalein (5,6,7-Trihydroxyflavone) and Wogonin (5,7-Dihydroxy-8-Methoxyflavone) Derivatives: Structural Modifications Inspired from Flavonoids in Scutellaria Baicalensis. Eur. J. Med. Chem. 2022, 243, 114733. [Google Scholar] [CrossRef]
  30. Moghaddam, E.; Teoh, B.-T.; Sam, S.-S.; Lani, R.; Hassandarvish, P.; Chik, Z.; Yueh, A.; Abubakar, S.; Zandi, K. Baicalin, a Metabolite of Baicalein with Antiviral Activity against Dengue Virus. Sci. Rep. 2015, 4, 5452. [Google Scholar] [CrossRef]
  31. Cathcart, M.-C.; Useckaite, Z.; Drakeford, C.; Semik, V.; Lysaght, J.; Gately, K.; O’Byrne, K.J.; Pidgeon, G.P. Anti-Cancer Effects of Baicalein in Non-Small Cell Lung Cancer in-Vitro and in-Vivo. BMC Cancer 2016, 16, 707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Li, K.; Li, J.; Xu, J.; Zhang, L.; Liu, Q.-Y.; Huang, Y.-K.; Zhang, Z.-G.; Kang, Y.; Xu, C.-J. Baicalein Ameliorates Chronic Stress-Mediated Ovarian Dysfunction by Upregulating the Expression of Gamma-Aminobutyric Acid B2 Receptor. Reprod. Dev. Med. 2018, 2, 21–29. [Google Scholar] [CrossRef]
  33. Takahashi, H.; Chen, M.C.; Pham, H.; Angst, E.; King, J.C.; Park, J.; Brovman, E.Y.; Ishiguro, H.; Harris, D.M.; Reber, H.A.; et al. Baicalein, a Component of Scutellaria Baicalensis, Induces Apoptosis by Mcl-1 down-Regulation in Human Pancreatic Cancer Cells. Biochim. Biophys. Acta Mol. Cell Res. 2011, 1813, 1465–1474. [Google Scholar] [CrossRef] [Green Version]
  34. Ruan, C.; Xiao, X.; Li, G. Microwave-Assisted Extraction Coupled with Countercurrent Chromatography for the Rapid Preparation of Flavonoids from Scutellaria Barbata D. Don. J. Sep. Sci. 2014, 37, 1364–1369. [Google Scholar] [CrossRef] [PubMed]
  35. Sui, X.; Han, X.; Chen, P.; Wu, Q.; Feng, J.; Duan, T.; Chen, X.; Pan, T.; Yan, L.; Jin, T.; et al. Baicalin Induces Apoptosis and Suppresses the Cell Cycle Progression of Lung Cancer Cells Through Downregulating Akt/MTOR Signaling Pathway. Front. Mol. Biosci. 2021, 7, 602282. [Google Scholar] [CrossRef] [PubMed]
  36. Tao, Y.; Zhan, S.; Wang, Y.; Zhou, G.; Liang, H.; Chen, X.; Shen, H. Baicalin, the Major Component of Traditional Chinese Medicine Scutellaria Baicalensis Induces Colon Cancer Cell Apoptosis through Inhibition of OncomiRNAs. Sci. Rep. 2018, 8, 14477. [Google Scholar] [CrossRef] [Green Version]
  37. Lu, Q.-Y.; Zhang, L.; Moro, A.; Chen, M.C.; Harris, D.M.; Eibl, G.; Go, V.-L.W. Detection of Baicalin Metabolites Baicalein and Oroxylin-A in Mouse Pancreas and Pancreatic Xenografts. Pancreas 2012, 41, 571–576. [Google Scholar] [CrossRef] [Green Version]
  38. Li, H.-B.; Chen, F. Isolation and Purification of Baicalein, Wogonin and Oroxylin A from the Medicinal Plant Scutellaria Baicalensis by High-Speed Counter-Current Chromatography. J. Chromatogr. A 2005, 1074, 107–110. [Google Scholar] [CrossRef]
  39. Lin, M.-Y.; Cheng, W.-T.; Cheng, H.-C.; Chou, W.-C.; Chen, H.-I.; Ou, H.-C.; Tsai, K.-L. Baicalin Enhances Chemosensitivity to Doxorubicin in Breast Cancer Cells via Upregulation of Oxidative Stress-Mediated Mitochondria-Dependent Apoptosis. Antioxidants 2021, 10, 1506. [Google Scholar] [CrossRef]
  40. Wang, S.; Fu, J.L.; Hao, H.F.; Jiao, Y.N.; Li, P.P.; Han, S.Y. Metabolic reprogramming by traditional Chinese medicine and its role in effective cancer therapy. Pharmacol. Res. 2021, 170, 105728. [Google Scholar] [CrossRef] [PubMed]
  41. Tian, Y.; Li, Q.; Zhou, X.; Pang, Q.; Xu, Y. A UHPLC–MS/MS Method for Simultaneous Determination of Twelve Constituents from Erigeron Breviscapus Extract in Rat Plasma: Application to a Pharmacokinetic Study. J. Chromatogr. B 2017, 1046, 1–12. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, Y.; Tan, L.; Jiao, K.; Xue, C.; Tang, Q.; Jiang, S.; Ren, Y.; Chen, H.; El-Aziz, T.M.A.; Abdelazeem, K.N.M.; et al. Scutellarein Attenuates Atopic Dermatitis by Selectively Inhibiting Transient Receptor Potential Vanilloid 3 Channels. Br. J. Pharmacol. 2022, 179, 4792–4808. [Google Scholar] [CrossRef] [PubMed]
  43. Gu, T.; Zhong, Y.; Lu, Y.-T.; Sun, Y.; Dong, Z.-X.; Wu, W.-Y.; Shi, Z.-H.; Li, N.-G.; Xue, X.; Fang, F.; et al. Synthesis and Bioactivity Characterization of Scutellarein Sulfonated Derivative. Molecules 2017, 22, 1028. [Google Scholar] [CrossRef] [Green Version]
  44. Wang, M.; Firrman, J.; Liu, L.; Yam, K. A Review on Flavonoid Apigenin: Dietary Intake, ADME, Antimicrobial Effects, and Interactions with Human Gut Microbiota. Biomed. Res. Int. 2019, 2019, 7010467. [Google Scholar] [CrossRef] [PubMed]
  45. Lu, J.; Meng, Z.; Chen, Y.; Yu, L.; Gao, B.; Zheng, Y.; Guan, S. Apigenin Induced Autophagy and Stimulated Autophagic Lipid Degradation. Food Funct. 2020, 11, 9208–9215. [Google Scholar] [CrossRef] [PubMed]
  46. Ni, G.; Tang, Y.; Li, M.; He, Y.; Rao, G. Synthesis of Scutellarein Derivatives with a Long Aliphatic Chain and Their Biological Evaluation against Human Cancer Cells. Molecules 2018, 23, 310. [Google Scholar] [CrossRef] [Green Version]
  47. Liu, K.; Tian, T.; Zheng, Y.; Zhou, L.; Dai, C.; Wang, M.; Lin, S.; Deng, Y.; Hao, Q.; Zhai, Z.; et al. Scutellarin Inhibits Proliferation and Invasion of Hepatocellular Carcinoma Cells via Down-regulation of JAK2/STAT3 Pathway. J. Cell. Mol. Med. 2019, 23, 3040–3044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Yuan, Y.; Fang, M.; Wu, C.-Y.; Ling, E.-A. Scutellarin as a Potential Therapeutic Agent for Microglia-Mediated Neuroinflammation in Cerebral Ischemia. Neuromolecular Med. 2016, 18, 264–273. [Google Scholar] [CrossRef] [PubMed]
  49. Gao, C.; Zhou, Y.; Jiang, Z.; Zhao, Y.; Zhang, D.; Cong, X.; Cao, R.; Li, H.; Tian, W. Cytotoxic and Chemosensitization Effects of Scutellarin from Traditional Chinese Herb Scutellaria Altissima L. in Human Prostate Cancer Cells. Oncol. Rep. 2017, 38, 1491–1499. [Google Scholar] [CrossRef] [Green Version]
  50. Yang, B.; Zhao, Y.-L.; Yang, X.; Liao, X.-L.; Yang, J.; Zhang, J.-H.; Gao, C.-Z. Scutellarin-Cyclodextrin Conjugates: Synthesis, Characterization and Anticancer Activity. Carbohydr. Polym. 2013, 92, 1308–1314. [Google Scholar] [CrossRef]
  51. Liu, Y.; Jing, Y.-Y.; Zeng, C.-Y.; Li, C.-G.; Xu, L.-H.; Yan, L.; Bai, W.-J.; Zha, Q.-B.; Ouyang, D.-Y.; He, X.-H. Scutellarin Suppresses NLRP3 Inflammasome Activation in Macrophages and Protects Mice against Bacterial Sepsis. Front. Pharmacol. 2018, 8, 975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Chan, E.W.C.; Lim, C.S.Q.; Lim, W.Y.; Loong, Z.J.; Wong, C.W. Role of Scutellarin in Human Cancer—A Review. J. Appl. Pharm. Sci. 2019, 9, 143–146. [Google Scholar]
  53. Hollman, P.C.H.; Bijsman, M.N.C.P.; van Gameren, Y.; Cnossen, E.P.J.; de Vries, J.H.M.; Katan, M.B. The Sugar Moiety Is a Major Determinant of the Absorption of Dietary Flavonoid Glycosides in Man. Free Radic. Res. 1999, 31, 569–573. [Google Scholar] [CrossRef] [PubMed]
  54. Xiao, J. Dietary Flavonoid Aglycones and Their Glycosides: Which Show Better Biological Significance? Crit. Rev. Food Sci. Nutr. 2015, 57, 1874–1905. [Google Scholar] [CrossRef]
  55. Naeem, A.; Ming, Y.; Pengyi, H.; Jie, K.Y.; Yali, L.; Haiyan, Z.; Shuai, X.; Wenjing, L.; Ling, W.; Xia, Z.M.; et al. The Fate of Flavonoids after Oral Administration: A Comprehensive Overview of Its Bioavailability. Crit. Rev. Food Sci. Nutr. 2022, 62, 6169–6186. [Google Scholar] [CrossRef]
  56. Day, A.J.; Cañada, F.J.; Dñaz, J.C.; Kroon, P.A.; Mclauchlan, R.; Faulds, C.B.; Plumb, G.W.; Morgan, M.R.A.; Williamson, G. Dietary Flavonoid and Isoflavone Glycosides Are Hydrolysed by the Lactase Site of Lactase Phlorizin Hydrolase. FEBS Lett. 2000, 468, 166–170. [Google Scholar] [CrossRef] [Green Version]
  57. Akhlaghi, M.; Foshati, S. Bioavailability and Metabolism of Flavonoids: A Review. Int. J. Nutr. Sci. 2017, 2, 180–184. [Google Scholar]
  58. Xu, J.; Zhang, B.; Chu, Z.; Jiang, F.; Han, J. Wogonin Alleviates Cisplatin-Induced Cardiotoxicity in Mice Via Inhibiting Gasdermin D-Mediated Pyroptosis. J. Cardiovasc. Pharmacol. 2021, 78, 597–603. [Google Scholar] [CrossRef]
  59. Wang, T.; Long, F.; Jiang, G.; Cai, H.; Jiang, Q.; Cheng, K.; Hu, Z.; Wang, Y. Pharmacokinetic Properties of Wogonin and Its Herb-Drug Interactions with Docetaxel in Rats with Mammary Tumors. Biomed. Chromatogr. 2018, 32, e4264. [Google Scholar] [CrossRef]
  60. Fong, S.Y.K.; Wong, Y.C.; Zuo, Z. Development of a SPE-LC/MS/MS Method for Simultaneous Quantification of Baicalein, Wogonin, Oroxylin A and Their Glucuronides Baicalin, Wogonoside and Oroxyloside in Rats and Its Application to Brain Uptake and Plasma Pharmacokinetic Studies. J. Pharm. Biomed. Anal. 2014, 97, 9–23. [Google Scholar] [CrossRef]
  61. Wang, Q.; Shi, R.; Dai, Y.; Li, Y.; Wang, T.; Ma, Y.; Cheng, N. Mechanism in the Existent Difference in Form of Wogonin/Wogonoside between Plasma and Intestine/Liver in Rats. RSC Adv. 2018, 8, 3364–3373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Talbi, A.; Zhao, D.; Liu, Q.; Li, J.; Fan, A.; Yang, W.; Han, X.; Chen, X. Pharmacokinetics, Tissue Distribution, Excretion and Plasma Protein Binding Studies of Wogonin in Rats. Molecules 2014, 19, 5538–5549. [Google Scholar] [CrossRef]
  63. Zhang, J.; Lv, H.; Jiang, K.; Gao, Y. Enhanced Bioavailability after Oral and Pulmonary Administration of Baicalein Nanocrystal. Int. J. Pharm. 2011, 420, 180–188. [Google Scholar] [CrossRef] [PubMed]
  64. Zhu, M.-L.; Liang, X.-L.; Zhao, L.-J.; Liao, Z.-G.; Zhao, G.-W.; Cao, Y.-C.; Zhang, J.; Luo, Y. Elucidation of the Transport Mechanism of Baicalin and the Influence of a Radix Angelicae Dahuricae Extract on the Absorption of Baicalin in a Caco-2 Cell Monolayer Model. J. Ethnopharmacol. 2013, 150, 553–559. [Google Scholar] [CrossRef] [PubMed]
  65. Liu, Z.; Zhang, X.; Li, J.; Liu, R.; Shu, L.; Jin, J. Effects of Labrasol on the Corneal Drug Delivery of Baicalin. Drug Deliv. 2009, 16, 399–404. [Google Scholar] [CrossRef]
  66. Taiming, L.; Xuehua, J. Investigation of the Absorption Mechanisms of Baicalin and Baicalein in Rats. J. Pharm. Sci. 2006, 95, 1326–1333. [Google Scholar] [CrossRef]
  67. Xing, J.; Chen, X.; Sun, Y.; Luan, Y.; Zhong, D. Interaction of Baicalin and Baicalein with Antibiotics in the Gastrointestinal Tract. J. Pharm. Pharmacol. 2010, 57, 743–750. [Google Scholar] [CrossRef]
  68. Lai, M.-Y.; Hsiu, S.-L.; Tsai, S.-Y.; Hou, Y.-C.; Chao, P.-D.L. Comparison of Metabolic Pharmacokinetics of Baicalin and Baicalein in Rats. J. Pharm. Pharmacol. 2010, 55, 205–209. [Google Scholar] [CrossRef]
  69. Wei, J.; Liu, R.; Zhang, J.; Liu, S.; Yan, D.; Wen, X.; Tian, X. Baicalin Enhanced Oral Bioavailability of Sorafenib in Rats by Inducing Intestine Absorption. Front. Pharmacol. 2021, 12. [Google Scholar] [CrossRef]
  70. Abe, K.; Inoue, O.; Yumioka, E. Biliary Excretion of Metabolites of Baicalin, and Baicalein in Rats. Chem. Pharm. Bull. 1990, 38, 208–211. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, L.; Lin, G.; Chang, Q.; Zuo, Z. Role of Intestinal First-Pass Metabolism of Baicalein in Its Absorption Process. Pharm. Res. 2005, 22, 1050–1058. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, J.-L.; Che, Q.-M.; Li, S.-Z.; Zhou, T.-H. Study on Metabolism of Scutellarin in Rats by HPLC-MS and HPLC-NMR. J. Asian Nat. Prod. Res. 2003, 5, 249–256. [Google Scholar] [CrossRef] [PubMed]
  73. Qiu, F.; Xia, H.; Zhang, T.; Di, X.; Qu, G.; Yao, X. Two Major Urinary Metabolites of Scutellarin in Rats. Planta Med. 2007, 73, 363–365. [Google Scholar] [CrossRef]
  74. Gao, H.-M.; Wang, Z.-M.; Tian, J. Pharmacokinetics and Metabolites of Scutellarin in Normal and Model Rats. Acta Pharm. Sin. 2005, 40, 1024–1027. [Google Scholar]
  75. Gao, C.; Zhang, H.; Guo, Z.; You, T.; Chen, X.; Zhong, D. Mechanistic Studies on the Absorption and Disposition of Scutellarin in Humans: Selective OATP2B1-Mediated Hepatic Uptake Is a Likely Key Determinant for Its Unique Pharmacokinetic Characteristics. Drug Metab. Dispos. 2012, 40, 2009–2020. [Google Scholar] [CrossRef] [Green Version]
  76. Gao, C.; Chen, X.; Zhong, D. Absorption and Disposition of Scutellarin in Rats: A Pharmacokinetic Explanation for the High Exposure of Its Isomeric Metabolite. Drug Metab. Dispos. 2011, 39, 2034–2044. [Google Scholar] [CrossRef]
  77. Patra, S.; Mishra, S.R.; Behera, B.P.; Mahapatra, K.K.; Panigrahi, D.P.; Bhol, C.S.; Praharaj, P.P.; Sethi, G.; Patra, S.K.; Bhutia, S.K. Autophagy-Modulating Phytochemicals in Cancer Therapeutics: Current Evidences and Future Perspectives. Semin. Cancer Biol. 2022, 80, 205–217. [Google Scholar] [CrossRef]
  78. Bhutia, S.K.; Behera, B.; Nandini Das, D.; Mukhopadhyay, S.; Sinha, N.; Panda, P.K.; Naik, P.P.; Patra, S.K.; Mandal, M.; Sarkar, S.; et al. Abrus Agglutinin Is a Potent Anti-Proliferative and Anti-Angiogenic Agent in Human Breast Cancer. Int. J. Cancer 2016, 139, 457–466. [Google Scholar] [CrossRef] [Green Version]
  79. Mathew, L.K.; Sengupta, S.; Kawakami, A.; Andreasen, E.A.; Löhr, C.V.; Loynes, C.A.; Renshaw, S.A.; Peterson, R.T.; Tanguay, R.L. Unraveling Tissue Regeneration Pathways Using Chemical Genetics. J. Biol. Chem. 2007, 282, 35202–35210. [Google Scholar] [CrossRef] [Green Version]
  80. Kanzawa, T.; Bedwell, J.; Kondo, Y.; Kondo, S.; Germano, I.M. Inhibition of DNA Repair for Sensitizing Resistant Glioma Cells to Temozolomide. J. Neurosurg. 2003, 99, 1047–1052. [Google Scholar] [CrossRef] [Green Version]
  81. Ostenfeld, M.S.; Fehrenbacher, N.; Høyer-Hansen, M.; Thomsen, C.; Farkas, T.; Jäättelä, M. Effective Tumor Cell Death by σ-2 Receptor Ligand Siramesine Involves Lysosomal Leakage and Oxidative Stress. Cancer Res. 2005, 65, 8975–8983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Fazi, B.; Bursch, W.; Fimia, G.M.; Nardacci, R.; Piacentini, M.; di Sano, F.; Piredda, L. Fenretinide Induces Autophagic Cell Death in Caspase-Defective Breast Cancer Cells. Autophagy 2008, 4, 435–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Grandér, D.; Kharaziha, P.; Laane, E.; Pokrovskaja, K.; Panaretakis, T. Autophagy as the Main Means of Cytotoxicity by Glucocorticoids in Hematological Malignancies. Autophagy 2009, 5, 1198–1200. [Google Scholar] [CrossRef] [PubMed]
  84. Laane, E.; Tamm, K.P.; Buentke, E.; Ito, K.; Khahariza, P.; Oscarsson, J.; Corcoran, M.; Björklund, A.-C.; Hultenby, K.; Lundin, J.; et al. Cell Death Induced by Dexamethasone in Lymphoid Leukemia Is Mediated through Initiation of Autophagy. Cell Death Differ. 2009, 16, 1018–1029. [Google Scholar] [CrossRef] [Green Version]
  85. Kim, D.-H.; Sarbassov, D.D.; Ali, S.M.; King, J.E.; Latek, R.R.; Erdjument-Bromage, H.; Tempst, P.; Sabatini, D.M. MTOR Interacts with Raptor to Form a Nutrient-Sensitive Complex That Signals to the Cell Growth Machinery. Cell 2002, 110, 163–175. [Google Scholar] [CrossRef] [Green Version]
  86. Shi, H.; Zhang, L.; Zhang, C.; Hao, Y.; Zhao, X. Rapamycin May Inhibit Murine S180 Sarcoma Growth by Regulating the Pathways Associated with Autophagy and Cancer Stem Cells. J. Cancer Res. Ther. 2019, 15, 398–403. [Google Scholar] [CrossRef]
  87. Liu, Y.-L.; Yang, P.-M.; Shun, C.-T.; Wu, M.-S.; Weng, J.-R.; Chen, C.-C. Autophagy Potentiates the Anti-Cancer Effects of the Histone Deacetylase Inhibitors in Hepatocellular Carcinoma. Autophagy 2010, 6, 1057–1065. [Google Scholar] [CrossRef] [Green Version]
  88. Lin, X.; Han, L.; Weng, J.; Wang, K.; Chen, T. Rapamycin Inhibits Proliferation and Induces Autophagy in Human Neuroblastoma Cells. Biosci. Rep. 2018, 38, BSR20181822. [Google Scholar] [CrossRef] [Green Version]
  89. Metibemu, D.S.; Akinloye, O.A.; Akamo, A.J.; Ojo, D.A.; Okeowo, O.T.; Omotuyi, I.O. Exploring Receptor Tyrosine Kinases-Inhibitors in Cancer Treatments. Egypt. J. Med. Hum. Genet. 2019, 20, 35. [Google Scholar] [CrossRef] [Green Version]
  90. Kondo, Y.; Kondo, S. Autophagy and Cancer Therapy. Autophagy 2006, 2, 85–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Liu, R.; Chen, Y.; Liu, G.; Li, C.; Song, Y.; Cao, Z.; Li, W.; Hu, J.; Lu, C.; Liu, Y. PI3K/AKT Pathway as a Key Link Modulates the Multidrug Resistance of Cancers. Cell Death Dis. 2020, 11, 797. [Google Scholar] [CrossRef] [PubMed]
  92. Filomeni, G.; de Zio, D.; Cecconi, F. Oxidative Stress and Autophagy: The Clash between Damage and Metabolic Needs. Cell Death Differ. 2015, 22, 377–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Poillet-Perez, L.; White, E. Role of Tumor and Host Autophagy in Cancer Metabolism. Genes Dev. 2019, 33, 610–619. [Google Scholar] [CrossRef] [PubMed]
  94. Chouaib, S.; Noman, M.Z.; Kosmatopoulos, K.; Curran, M.A. Hypoxic Stress: Obstacles and Opportunities for Innovative Immunotherapy of Cancer. Oncogene 2017, 36, 439–445. [Google Scholar] [CrossRef] [Green Version]
  95. Paglin, S.; Hollister, T.; Delohery, T.; Hackett, N.; McMahill, M.; Sphicas, E.; Domingo, D.; Yahalom, J. A Novel Response of Cancer Cells to Radiation Involves Autophagy and Formation of Acidic Vesicles. Cancer Res. 2001, 61, 439–444. [Google Scholar]
  96. Liang, C.; Feng, P.; Ku, B.; Dotan, I.; Canaani, D.; Oh, B.-H.; Jung, J.U. Autophagic and Tumour Suppressor Activity of a Novel Beclin1-Binding Protein UVRAG. Nat. Cell Biol. 2006, 8, 688–698. [Google Scholar] [CrossRef]
  97. Hoare, M.; Young, A.R.J.; Narita, M. Autophagy in Cancer: Having Your Cake and Eating It. Semin. Cancer Biol. 2011, 21, 397–404. [Google Scholar] [CrossRef]
  98. White, E.; DiPaola, R.S. The Double-Edged Sword of Autophagy Modulation in Cancer. Clin. Cancer Res. 2009, 15, 5308–5316. [Google Scholar] [CrossRef] [Green Version]
  99. Chen, N.; Karantza-Wadsworth, V. Role and Regulation of Autophagy in Cancer. Biochim. Biophys. Acta Mol. Cell Res. 2009, 1793, 1516–1523. [Google Scholar] [CrossRef] [Green Version]
  100. Sikder, S.; Kumari, S.; Mustafi, P.; Ramdas, N.; Padhi, S.; Saha, A.; Bhaduri, U.; Banerjee, B.; Manjithaya, R.; Kundu, T.K. Nonhistone Human Chromatin Protein PC4 Is Critical for Genomic Integrity and Negatively Regulates Autophagy. FEBS J. 2019, 286, 4422–4442. [Google Scholar] [CrossRef]
  101. Maclean, K.H.; Dorsey, F.C.; Cleveland, J.L.; Kastan, M.B. Targeting Lysosomal Degradation Induces P53-Dependent Cell Death and Prevents Cancer in Mouse Models of Lymphomagenesis. J. Clin. Investig. 2008, 118, 79–88. [Google Scholar] [CrossRef] [Green Version]
  102. Liang, X.; Tang, J.; Liang, Y.; Jin, R.; Cai, X. Suppression of Autophagy by Chloroquine Sensitizes 5-Fluorouracil-Mediated Cell Death in Gallbladder Carcinoma Cells. Cell Biosci. 2014, 4, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Boya, P.; González-Polo, R.-A.; Casares, N.; Perfettini, J.-L.; Dessen, P.; Larochette, N.; Métivier, D.; Meley, D.; Souquere, S.; Yoshimori, T.; et al. Inhibition of Macroautophagy Triggers Apoptosis. Mol. Cell. Biol. 2005, 25, 1025–1040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Amaravadi, R.K.; Yu, D.; Lum, J.J.; Bui, T.; Christophorou, M.A.; Evan, G.I.; Thomas-Tikhonenko, A.; Thompson, C.B. Autophagy Inhibition Enhances Therapy-Induced Apoptosis in a Myc-Induced Model of Lymphoma. J. Clin. Investig. 2007, 117, 326–336. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, S.; Rehman, S.K.; Zhang, W.; Wen, A.; Yao, L.; Zhang, J. Autophagy Is a Therapeutic Target in Anticancer Drug Resistance. Biochim. Biophys. Acta Rev. Cancer 2010, 1806, 220–229. [Google Scholar] [CrossRef] [PubMed]
  106. Deldar Abad Paskeh, M.; Mirzaei, S.; Ashrafizadeh, M.; Zarrabi, A.; Sethi, G. Wnt/β-Catenin Signaling as a Driver of Hepatocellular Carcinoma Progression: An Emphasis on Molecular Pathways. J. Hepatocell. Carcinoma 2021, 8, 1415–1444. [Google Scholar] [CrossRef]
  107. Ong, P.S.; Wang, L.Z.; Dai, X.; Tseng, S.H.; Loo, S.J.; Sethi, G. Judicious Toggling of MTOR Activity to Combat Insulin Resistance and Cancer: Current Evidence and Perspectives. Front. Pharmacol. 2016, 7, 395. [Google Scholar] [CrossRef] [Green Version]
  108. Lampada, A.; O’Prey, J.; Szabadkai, G.; Ryan, K.M.; Hochhauser, D.; Salomoni, P. MTORC1-Independent Autophagy Regulates Receptor Tyrosine Kinase Phosphorylation in Colorectal Cancer Cells via an MTORC2-Mediated Mechanism. Cell Death Differ. 2017, 24, 1045–1062. [Google Scholar] [CrossRef] [Green Version]
  109. Alsagaby, S.A.; Iqbal, D.; Ahmad, I.; Patel, H.; Mir, S.A.; Madkhali, Y.A.; Oyouni, A.A.A.; Hawsawi, Y.M.; Alhumaydhi, F.A.; Alshehri, B.; et al. In Silico Investigations Identified Butyl Xanalterate to Competently Target CK2α (CSNK2A1) for Therapy of Chronic Lymphocytic Leukemia. Sci. Rep. 2022, 12, 17648. [Google Scholar] [CrossRef]
  110. Pottier, C.; Fresnais, M.; Gilon, M.; Jérusalem, G.; Longuespée, R.; Sounni, N.E. Tyrosine Kinase Inhibitors in Cancer: Breakthrough and Challenges of Targeted Therapy. Cancers 2020, 12, 731. [Google Scholar] [CrossRef] [Green Version]
  111. Arora, A.; Scholar, E.M. Role of Tyrosine Kinase Inhibitors in Cancer Therapy. J. Pharmacol. Exp. Ther. 2005, 315, 971–979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kumar, R.; Harilal, S.; Parambi, D.G.T.; Narayanan, S.E.; Uddin, M.S.; Marathakam, A.; Jose, J.; Mathew, G.E.; Mathew, B. Fascinating Chemopreventive Story of Wogonin: A Chance to Hit on the Head in Cancer Treatment. Curr. Pharm. Des. 2021, 27, 467–478. [Google Scholar] [CrossRef] [PubMed]
  113. Sharifi-Rad, J.; Herrera-Bravo, J.; Salazar, L.A.; Shaheen, S.; Abdulmajid Ayatollahi, S.; Kobarfard, F.; Imran, M.; Imran, A.; Custódio, L.; Dolores López, M.; et al. The Therapeutic Potential of Wogonin Observed in Preclinical Studies. Evid. Based Complement. Altern. Med. 2021, 2021, 9935451. [Google Scholar] [CrossRef] [PubMed]
  114. Law, B.Y.K.; Chan, W.K.; Xu, S.W.; Wang, J.R.; Bai, L.P.; Liu, L.; Wong, V.K.W. Natural Small-Molecule Enhancers of Autophagy Induce Autophagic Cell Death in Apoptosis-Defective Cells. Sci. Rep. 2014, 4, 5510. [Google Scholar] [CrossRef]
  115. Wu, K.; Teng, M.; Zhou, W.; Lu, F.; Zhou, Y.; Zeng, J.; Yang, J.; Liu, X.; Zhang, Y.; Ding, Y.; et al. Wogonin Induces Cell Cycle Arrest and Apoptosis of Hepatocellular Carcinoma Cells by Activating Hippo Signaling. Anticancer Agents Med. Chem. 2022, 22, 1551–1560. [Google Scholar] [CrossRef]
  116. Chen, M.; Wu, H.L.; Wong, T.S.; Chen, B.; Gong, R.-H.; Wong, H.L.X.; Xiao, H.; Bian, Z.; Kwan, H.Y. Combination of Wogonin and Artesunate Exhibits Synergistic Anti-Hepatocellular Carcinoma Effect by Increasing DNA-Damage-Inducible Alpha, Tumor Necrosis Factor α and Tumor Necrosis Factor Receptor-Associated Factor 3-Mediated Apoptosis. Front. Pharmacol. 2021, 12. [Google Scholar] [CrossRef]
  117. Cui, B.; Yu, J.-M. Autophagy: A New Pathway for Traditional Chinese Medicine. J. Asian Nat. Prod. Res. 2018, 20, 14–26. [Google Scholar] [CrossRef]
  118. Zhu, Y.; Liu, M.; Yao, J.; Guo, Q.; Wei, L. The Synthetic Flavonoid Derivative GL-V9 Induces Apoptosis and Autophagy in Cutaneous Squamous Cell Carcinoma via Suppressing AKT-Regulated HK2 and MTOR Signals. Molecules 2020, 25, 5033. [Google Scholar] [CrossRef]
  119. Li, S.-J.; Sun, S.-J.; Gao, J.; Sun, F.-B. Wogonin Induces Beclin-1/PI3K and Reactive Oxygen Species-Mediated Autophagy in Human Pancreatic Cancer Cells. Oncol. Lett. 2016, 12, 5059–5067. [Google Scholar] [CrossRef] [Green Version]
  120. Hong, Z.-P.; Wang, L.-G.; Wang, H.-J.; Ye, W.-F.; Wang, X.-Z. Wogonin Exacerbates the Cytotoxic Effect of Oxaliplatin by Inducing Nitrosative Stress and Autophagy in Human Gastric Cancer Cells. Phytomedicine 2018, 39, 168–175. [Google Scholar] [CrossRef]
  121. Zhang, H.; Ge, C.; Lin, C.; Yi, L.; Ran, J.; Shi, X.; Tang, C.; Wu, Y.; Nian, W. Combination of Icotinib and Wogonin Induces Apoptosis and Autophagy to Overcome Acquired Resistance in Lung Cancer Harbouring EGFR T790M Mutation. Int. J. Clin. Exp. Med. 2017, 10, 7553–7562. [Google Scholar]
  122. Huang, S.T.; Wang, C.Y.; Yang, R.C.; Chu, C.J.; Wu, H.T.; Pang, J.H. Wogonin, an active compound in Scutellaria baicalensis, induces apoptosis and reduces telomerase activity in the HL-60 leukemia cells. Phytomedicine 2010, 17, 47–54. [Google Scholar] [CrossRef] [PubMed]
  123. Ma, L.; Wu, F.; Shao, Q.; Chen, G.; Xu, L.; Lu, F. Baicalin Alleviates Oxidative Stress and Inflammation in Diabetic Nephropathy via Nrf2 and MAPK Signaling Pathway. Drug Des. Devel. Ther. 2021, 15, 3207–3221. [Google Scholar] [CrossRef] [PubMed]
  124. Pan, L.; Cho, K.-S.; Yi, I.; To, C.-H.; Chen, D.F.; Do, C.-W. Baicalein, Baicalin, and Wogonin: Protective Effects against Ischemia-Induced Neurodegeneration in the Brain and Retina. Oxid. Med. Cell. Longev. 2021, 2021, 8377362. [Google Scholar] [CrossRef] [PubMed]
  125. Zhu, Y.; Fang, J.; Wang, H.; Fei, M.; Tang, T.; Liu, K.; Niu, W.; Zhou, Y. Baicalin Suppresses Proliferation, Migration, and Invasion in Human Glioblastoma Cells via Ca2+-Dependent Pathway. Drug Des. Devel. Ther. 2018, 12, 3247–3261. [Google Scholar] [CrossRef] [PubMed]
  126. Gong, W.; Zhao, Z.; Liu, B.; Lu, L.; Dong, J. Exploring the Chemopreventive Properties and Perspectives of Baicalin and Its Aglycone Baicalein in Solid Tumors. Eur. J. Med. Chem. 2017, 126, 844–852. [Google Scholar] [CrossRef]
  127. Wang, B.; Huang, T.; Fang, Q.; Zhang, X.; Yuan, J.; Li, M.; Ge, H. Bone-Protective and Anti-Tumor Effect of Baicalin in Osteotropic Breast Cancer via Induction of Apoptosis. Breast Cancer Res. Treat. 2020, 184, 711–721. [Google Scholar] [CrossRef]
  128. Lan, H.; Wang, H.; Gao, M.; Luo, G.; Zhang, J.; Yi, E.; Liang, C.; Xiong, X.; Chen, X.; Wu, Q.; et al. Analysis and Construction of a Competitive Endogenous RNA Regulatory Network of Baicalin-Induced Apoptosis in Human Osteosarcoma Cells. Biomed. Res. Int. 2021, 2021, 9984112. [Google Scholar] [CrossRef]
  129. Liu, Y.; Hong, Z.; Chen, P.; Wang, J.; Zhou, Y.; Huang, J. Baicalin Inhibits Growth and Induces Apoptosis of Human Osteosarcoma Cells by Suppressing the AKT Pathway. Oncol. Lett. 2019, 18, 3188–3194. [Google Scholar] [CrossRef] [Green Version]
  130. Wan, D.; Ouyang, H. Baicalin Induces Apoptosis in Human Osteosarcoma Cell through ROS-Mediated Mitochondrial Pathway. Nat. Prod. Res. 2018, 32, 1996–2000. [Google Scholar] [CrossRef]
  131. Wang, Y.; Wang, H.; Zhou, R.; Zhong, W.; Lu, S.; Ma, Z.; Chai, Y. Baicalin Inhibits Human Osteosarcoma Cells Invasion, Metastasis, and Anoikis Resistance by Suppressing the Transforming Growth Factor-Β1-Induced Epithelial-to-Mesenchymal Transition. Anticancer Drugs 2017, 28, 581–587. [Google Scholar] [CrossRef] [PubMed]
  132. Aryal, P.; Kim, K.; Park, P.-H.; Ham, S.; Cho, J.; Song, K. Baicalein Induces Autophagic Cell Death through AMPK/ULK1 Activation and Downregulation of MTORC1 Complex Components in Human Cancer Cells. FEBS J. 2014, 281, 4644–4658. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, Y.-F.; Xu, Y.-L.; Tang, Z.-H.; Li, T.; Zhang, L.-L.; Chen, X.; Lu, J.-H.; Leung, C.-H.; Ma, D.-L.; Qiang, W.-A.; et al. Baicalein Induces Beclin 1- and Extracellular Signal-Regulated Kinase-Dependent Autophagy in Ovarian Cancer Cells. Am. J. Chin. Med. 2017, 45, 123–136. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, Z.; Jiang, C.; Chen, W.; Zhang, G.; Luo, D.; Cao, Y.; Wu, J.; Ding, Y.; Liu, B. Baicalein Induces Apoptosis and Autophagy via Endoplasmic Reticulum Stress in Hepatocellular Carcinoma Cells. Biomed. Res. Int. 2014, 2014, 732516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Deng, X.; Liu, J.; Liu, L.; Sun, X.; Huang, J.; Dong, J. Drp1-Mediated Mitochondrial Fission Contributes to Baicalein-Induced Apoptosis and Autophagy in Lung Cancer via Activation of AMPK Signaling Pathway. Int. J. Biol. Sci. 2020, 16, 1403–1416. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, M.; Qiu, S.; Qin, J. Baicalein Induced Apoptosis and Autophagy of Undifferentiated Thyroid Cancer Cells by the ERK/PI3K/Akt Pathway. Am. J. Transl. Res. 2019, 11, 3341–3352. [Google Scholar]
  137. Hu, J.; Wang, R.; Liu, Y.; Zhou, J.; Shen, K.; Dai, Y. Baicalein Represses Cervical Cancer Cell Growth, Cell Cycle Progression and Promotes Apoptosis via Blocking AKT/mTOR Pathway by the Regulation of circHIAT1/miR-19a-3p Axis. Onco. Targets Ther. 2021, 14, 905–916. [Google Scholar] [CrossRef]
  138. Fattahi, S.; Amjadi-Moheb, F.; Tabaripour, R. PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci. 2020, 262, 118513. [Google Scholar] [CrossRef] [PubMed]
  139. Li, P.; Hu, J.; Shi, B.; Tie, J. Baicalein Enhanced Cisplatin Sensitivity of Gastric Cancer Cells by Inducing Cell Apoptosis and Autophagy via Akt/MTOR and Nrf2/Keap 1 Pathway. Biochem. Biophys. Res. Commun. 2020, 531, 320–327. [Google Scholar] [CrossRef]
  140. Pang, H.; Wu, T.; Peng, Z.; Tan, Q.; Peng, X.; Zhan, Z.; Song, L.; Wei, B. Baicalin Induces Apoptosis and Autophagy in Human Osteosarcoma Cells by Increasing ROS to Inhibit PI3K/Akt/MTOR, ERK1/2 and β-Catenin Signaling Pathways. J. Bone Oncol. 2022, 33, 100415. [Google Scholar] [CrossRef]
  141. Wei, Z.; Chen, J.; Zuo, F.; Guo, J.; Sun, X.; Liu, D.; Liu, C. Traditional Chinese Medicine Has Great Potential as Candidate Drugs for Lung Cancer: A Review. J. Ethnopharmacol. 2023, 300, 115748. [Google Scholar] [CrossRef]
  142. Wang, L.; Ma, Q. Clinical Benefits and Pharmacology of Scutellarin: A Comprehensive Review. Pharmacol. Ther. 2018, 190, 105–127. [Google Scholar] [CrossRef] [PubMed]
  143. Birundadevi, M.; Sivashankar, R.; Mathukumar, S. Scutellarein Apoptosis Mediated by Mitochondria in Oral Squamous Cell Carcinomas. Biomed. Biotechnol. Res. J. 2022, 6, 60. [Google Scholar] [CrossRef]
  144. Tan, H.; Li, X.; Yang, W.H.; Kang, Y. A Flavone, Wogonin from Scutellaria Baicalensis Inhibits the Proliferation of Human Colorectal Cancer Cells by Inducing of Autophagy, Apoptosis and G2/M Cell Cycle Arrest via Modulating the PI3K/AKT and STAT3 Signalling Pathways. J. BUON 2019, 24, 1143–1149. [Google Scholar] [PubMed]
  145. Zeng, S.; Tan, L.; Sun, Q.; Chen, L.; Zhao, H.; Liu, M.; Yang, H.; Ren, S.; Ming, T.; Tang, S.; et al. Suppression of Colitis-Associated Colorectal Cancer by Scutellarin through Inhibiting Hedgehog Signaling Pathway Activity. Phytomedicine 2022, 98, 153972. [Google Scholar] [CrossRef] [PubMed]
  146. Sun, C.; Li, C.; Li, X.; Zhu, Y.; Su, Z.; Wang, X.; He, Q.; Zheng, G.; Feng, B. Scutellarin Induces Apoptosis and Autophagy in NSCLC Cells through ERK1/2 and AKT Signaling Pathways in Vitro and in Vivo. J. Cancer 2018, 9, 3247–3256. [Google Scholar] [CrossRef]
  147. Sun, C.-Y.; Nie, J.; Zheng, Z.-L.; Zhao, J.; Wu, L.-M.; Zhu, Y.; Su, Z.-Q.; Zheng, G.-J.; Feng, B. Renoprotective Effect of Scutellarin on Cisplatin-Induced Renal Injury in Mice: Impact on Inflammation, Apoptosis, and Autophagy. Biomed. Pharmacother. 2019, 112, 108647. [Google Scholar] [CrossRef]
  148. Shi, X.; Chen, G.; Liu, X.; Qiu, Y.; Yang, S.; Zhang, Y.; Fang, X.; Zhang, C.; Liu, X. Scutellarein Inhibits Cancer Cell Metastasis in Vitro and Attenuates the Development of Fibrosarcoma in Vivo. Int. J. Mol. Med. 2015, 35, 31–38. [Google Scholar] [CrossRef] [Green Version]
  149. Ha, S.E.; Kim, S.M.; Vetrivel, P.; Kim, H.H.; Bhosale, P.B.; Heo, J.D.; Lee, H.J.; Kim, G.S. Inhibition of Cell Proliferation and Metastasis by Scutellarein Regulating PI3K/Akt/NF-ΚB Signaling through PTEN Activation in Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 8841. [Google Scholar] [CrossRef]
  150. Nie, J.; Yang, H.-M.; Sun, C.-Y.; Liu, Y.-L.; Zhuo, J.-Y.; Zhang, Z.-B.; Lai, X.-P.; Su, Z.-R.; Li, Y.-C. Scutellarin Enhances Antitumor Effects and Attenuates the Toxicity of Bleomycin in H22 Ascites Tumor-Bearing Mice. Front. Pharmacol. 2018, 9. [Google Scholar] [CrossRef]
  151. Hou, L.; Chen, L.; Fang, L. Scutellarin Inhibits Proliferation, Invasion, and Tumorigenicity in Human Breast Cancer Cells by Regulating HIPPO-YAP Signaling Pathway. Med. Sci. Monit. 2017, 23, 5130–5138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Huang, X.; Xu, Y.; Sui, X.; Lin, H.; Xu, J.; Han, D.; Ye, D.; Lv, G.; Liu, Y.; Qu, X.; et al. Scutellarein Suppresses Aβ-induced Memory Impairment via Inhibition of the NF-κB Pathway in Vivo and in Vitro. Oncol. Lett. 2019, 17, 5581–5589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Wang, F.; Yang, B.; Zhao, Y.; Liao, X.; Gao, C.; Jiang, R.; Han, B.; Yang, J.; Liu, M.; Zhou, R. Host-Guest Inclusion System of Scutellarein with 2-Hydroxypropyl-Beta-Cyclodextrin: Preparation, Characterization, and Anticancer Activity. J. Biomater. Sci. Polym. Ed. 2014, 25, 594–607. [Google Scholar] [CrossRef] [PubMed]
  154. Saralamma, V.V.; Vetrivel, P.; Lee, H.; Kim, S.; Ha, S.; Murugesan, R.; Kim, E.; Heo, J.; Kim, G. Comparative Proteomic Analysis Uncovers Potential Biomarkers Involved in the Anticancer Effect of Scutellarein in Human Gastric Cancer Cells. Oncol. Rep. 2020, 44, 939–958. [Google Scholar] [CrossRef]
  155. Lang, X.; Chen, Z.; Yang, X.; Yan, Q.; Xu, M.; Liu, W.; He, Q.; Zhang, Y.; Cheng, W.; Zhao, W. Scutellarein Induces Apoptosis and Inhibits Proliferation, Migration, and Invasion in Ovarian Cancer via Inhibition of EZH2/FOXO1 Signaling. J. Biochem. Mol. Toxicol. 2021, 35, e22870. [Google Scholar] [CrossRef]
  156. Sang Eun, H.; Seong Min, K.; Ho Jeong, L.; Vetrivel, P.; Venkatarame Gowda Saralamma, V.; Jeong Doo, H.; Eun Hee, K.; Sang Joon, L.; Gon Sup, K. Scutellarein Induces Fas-Mediated Extrinsic Apoptosis and G2/M Cell Cycle Arrest in Hep3B Hepatocellular Carcinoma Cells. Nutrients 2019, 11, 263. [Google Scholar] [CrossRef]
  157. Yang, N.; Zhao, Y.; Wang, Z.; Liu, Y.; Zhang, Y. Scutellarin Suppresses Growth and Causes Apoptosis of Human Colorectal Cancer Cells by Regulating the P53 Pathway. Mol. Med. Rep. 2017, 15, 929–935. [Google Scholar] [CrossRef] [Green Version]
  158. Chow, S.-E.; Chen, Y.-W.; Liang, C.-A.; Huang, Y.-K.; Wang, J.-S. Wogonin Induces Cross-Regulation between Autophagy and Apoptosis via a Variety of Akt Pathway in Human Nasopharyngeal Carcinoma Cells. J. Cell. Biochem. 2012, 113, 3476–3485. [Google Scholar] [CrossRef]
  159. Yan, W.; Ma, X.; Zhao, X.; Zhang, S. Baicalein Induces Apoptosis and Autophagy of Breast Cancer Cells via Inhibiting PI3K/AKT Pathway in Vivo and Vitro. Drug Des. Devel. Ther. 2018, 12, 3961–3972. [Google Scholar] [CrossRef] [Green Version]
  160. Liu, B.; Ding, L.; Zhang, L.; Wang, S.; Wang, Y.; Wang, B.; Li, L. Baicalein Induces Autophagy and Apoptosis through AMPK Pathway in Human Glioma Cells. Am. J. Chin. Med. 2019, 47, 1405–1418. [Google Scholar] [CrossRef]
  161. Qiao, D.; Li, Y.; Xing, J.; Sun, P.; Wang, Y.; Zhang, Y.; Chen, L.; Ren, X.; Lin, Z.; Jin, J.; et al. Baicalein Inhibits PI3K/AKT Signaling Pathway and Induces Autophagy of MGC-803 Cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2019, 35, 613–618. [Google Scholar] [PubMed]
  162. Yi, S.; Liu, G.; Wu, Y.; Liang, Q.; Li, L. Baicalein Suppresses the Growth of the Human Thyroid Cancer Cells by Inducing Mitotic Catastrophe, Apoptosis and Autophagy via NF-KB Signalling Pathway. J. BUON 2020, 25, 389–394. [Google Scholar] [PubMed]
  163. Lin, C.; Tsai, S.-C.; Tseng, M.T.; Peng, S.-F.; Kuo, S.-C.; Lin, M.-W.; Hsu, Y.-M.; Lee, M.-R.; Amagaya, S.; Huang, W.-W.; et al. AKT Serine/Threonine Protein Kinase Modulates Baicalin-Triggered Autophagy in Human Bladder Cancer T24 Cells. Int. J. Oncol. 2013, 42, 993–1000. [Google Scholar] [CrossRef] [Green Version]
  164. Zhang, X.; Tang, X.; Liu, H.; Li, L.; Hou, Q.; Gao, J. Autophagy Induced by Baicalin Involves Downregulation of CD147 in SMMC-7721 Cells in Vitro. Oncol. Rep. 2012, 27, 1128–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Sun, C.-Y.; Zhu, Y.; Li, X.-F.; Wang, X.-Q.; Tang, L.-P.; Su, Z.-Q.; Li, C.-Y.; Zheng, G.-J.; Feng, B. Scutellarin Increases Cisplatin-Induced Apoptosis and Autophagy to Overcome Cisplatin Resistance in Non-Small Cell Lung Cancer via ERK/P53 and c-Met/AKT Signaling Pathways. Front. Pharmacol. 2018, 9, 92. [Google Scholar] [CrossRef]
Figure 1. The most important flavone constituents of Scutellaria plants.
Figure 1. The most important flavone constituents of Scutellaria plants.
Pharmaceuticals 16 00302 g001
Figure 2. Schematic diagram showing the role of Scutellaria flavonoes in the tumor environment.
Figure 2. Schematic diagram showing the role of Scutellaria flavonoes in the tumor environment.
Pharmaceuticals 16 00302 g002
Table 1. An overview of the main features of wogonin, baicalein, baicalin, scutellarein and scutellarin.
Table 1. An overview of the main features of wogonin, baicalein, baicalin, scutellarein and scutellarin.
CompoundSource Plant/PlantsBioactivitiesReference
WogoninScutellaria baicalensis Georgi radix, Andrographis paniculata Burm.f, Anodendron affine (Hook. & Arn.) Druce, Tetracera indica L.Antioxidant, antiviral, anti-inflammatory, antiproliferative[26,27,28]
Baicalein S baicalensis, S lateriflora, Oroxylum indicumPositive allosteric modulator of GABAA receptor, anxiolytic activity[29,30,31,32]
Baicalin S. lateriflora, S. galericulata, Thalictrum baicalense, Radix scutellariaeNeuroprotective, cardioprotective, antioxidant, antiatherosclerotic, antibacterial, anticoronaviral[37,38]
Scutellarein S lateriflora, Scoparia dulcis, Artemisia douglasiana, Asplenium belangeri (Fern)Anticancer, antiproliferative, antioxidant[41,42,43,52]
ScutellarinScutellaria spp., Erigeron spp.Liver diseases, cerebrovascular diseases, hyperlipidemia[46,51]
Table 2. Pharmacokinetic data of Scutellaria flavones.
Table 2. Pharmacokinetic data of Scutellaria flavones.
Compound Administration of CompoundPharmacokinetic AnalysisReference
Wogonin (W.O.)Oral administration in C57BL/6 mice
H9c2 cells
Modulated Gasdermin D protein in H9c2 cells; Attenuated CDDP-induced cardiotoxicity and showed antipyroptotic effects[58]
Oral administration in rats at 10, 20 and 40 mg/kg Modulate the activities of CYPs, P-gp and Cmax AUC0-t of W.O. were proportionally increased[59]
Oral administration of R.S. extract (300 mg/kg) to Sprague–Dawley ratsW.O. showed the ability to cross the blood–brain barrier [60]
I.G. administration of W.O. in ratsMetabolized/detected in the small intestine and liver[61]
I.V. WO dose i.e., 10, 20 and 40 mg/kg
I.G. 100 mg/kg dose
I.V. WO (20 mg/kg) in Sprague–Dawley rats
W.O. was detected in all examined tissues; the highest levels were found in the kidney and liver, and 21% was excreted as an unchanged drug[62]
BaicaleinOral administration (121 mg/kg bw) and Pulmonary administration (10 mg/kg) in Male Sprague–Dawley (S.D.) ratsOral baicalein nanocrystals:
Bioavailability of baicalein is 1.67-fold, showing rapid and extensive absorption
[63]
Oral administration Male SD rats 30 mg/kg baicaleinDistributed rapidly within 0.25 h and accumulated in the lung and liver
Quickly absorbed in plasma lung > kidney >liver
having T1/2z 8.08
[64]
Oral administration in normal rats (65 mg/kg)Baicalein was significantly higher in the stomach > liver > intestine [65]
In situ perfusion in Male Wistar ratsBaicalein was moderately absorbed as per the stomach > small intestine >colon[66]
BaicalinI.V. administration (230–250 g) of 37 µmol/kg to Male Wistar rats
Oral administration of 227 µmol/kg
T ½ = 0.12 ± 0.02 in I.V. administered rats in plasma. Plasma concentration of baicalin displayed a second peak over the 8 –12 h (i.v.) [67]
I.V and oral administration in Male Sprague–Dawley ratsRapid absorption and simultaneous glucuronidation/sulfation. The absorption rate was slower and the Cmax was lower for oral baicalin compared with I.V baicalein[68]
I.G. and oral administration in rats at a dose of 160 mg/kg Coadministration significantly upregulated the Cmax, AUC0-t, and AUC0-∞ of oral dose by 2.02, 1.65, 1.66-fold in male rats [69]
Oral Administration to Male rats Baicalein is significantly hydrolyzed in the gastrointestinal tract.
The total cumulative amounts of baicalin were 54% of the doses
[70]
Intestinal perfusion model: In rat in situ single-pass In the rat’s intestinal regions, baicalin underwent considerable metabolism via conjugative processes[71]
Scutellarin/
Scutellarein
Oral administration 200 mg kg/bw in Male Wistar rats Scutellarein is obtained after metabolizing scutellarin in the blood by glucuronic acid and methylating enzymes in the liver[72]
Oral administration of 80 mg kg/bw to Male Wistar rats Two metabolites of Scutellarin, viz., Scutellarein 6,7-di-o-β-D-glucuronide and Scutellarein observed in urine[73]
I.V administration of 36 mg kg/bw to ratsTotal of four metabolites were observed in the plasma. Scutellarin was metabolized via dehydroxylation and methylation[74]
Oral administration in human subjectGlucuronidation of scutellarin is mediated by uridine 5’-diphosphoglucuronosyltransferase (UGT) in rats and humans [75]
Scutellarin (S-7-G) at a dose of 75 mg/kg orally to Male Sprague–Dawley ratsS-7-G and S-6-G were spotted in the systemic circulation
S-7-G absorbed as aglycone after hydrolyzed in the intestinal Glucuronidation of S-7-G occurs in liver microsomes of rat
[76]
Abbreviations: IV: Intravenous; IG: Intragasteric; CDP: cresyl diphenyl phosphate; µmol: micro mole; S7G: Scutellarin; SD: Sprague–Dawley; UGT: uridine 5’-diphosphoglucuronosyltransferase; CYPs: Cytochrome P450s.
Table 3. Regulation of autophagic machinery by Scutellaria flavones in different cancer types.
Table 3. Regulation of autophagic machinery by Scutellaria flavones in different cancer types.
Scutellaria FlavonesType of CancerCell LineMechanisms of ActionRef.
WogoninPancreatic cancer Panc-1 and Colo-357↑ROS, Beclin-1/PI3K[119]
Wogonin and oxaliplatinGastric cancer BGC-823 cells↑phospho-JNK (Thr183/Tyr185), phospho-ULK1 (Ser555), ↑LC3II[120]
WogoninNasopharyngeal cancerNPC-TW076 and NPCTW039↑LC3 I/II cleavage, autophagosome/autolysosome, ↓Raf/ERK [158]
WogoninColorectal cancer SW1417, SW48, DLD-1, HCT-15, LS-180 and CCD-18Co↓AKT and STAT3, ↑Beclin-1 caspases 3/8/9 and Bax expressions[12]
Wogonin derivative GL-V9Cutaneous squamous cancerA431 cells↓Akt/mTOR pathway[118]
BaicaleinBreast cancerMCF-7 and MDA-MB-231↓PI3K/AKT, NF-κB[159]
BaicaleinGlioma cellsU251 cells↑LC3, ↑caspase-3, ↑phosphorylation of AMPK[160]
BaicaleinLung cancerA549 and H1299↑activated A.M.P.K., ↑Drp1-mediated mitochondrial fission[135]
BaicaleinOvarian cancerHEY and A2780 ↑LC3-II, ↑PARP, ↑phosphorylation of ERK, ↑Beclin-1[133]
BaicaleinHepatocellular cancerSMMC-7721 and Bel-7402↑endoplasmic reticulum (E.R.) stress, ↓Bcl-2, ↑J.N.K.[153]
BaicaleinThyroid cancerF.R.O.↓Bcl-2/Bax, ↑Caspase-3, ↑Caspase-8, ↑Beclin-1, Atg5, ↓ERK, ↓PI3K/Akt [136]
BaicaleinGastric cancerMGC-803↑LC3, ↓PI3K and ↓AKT, ↑P62 [161]
BaicaleinProstate and breast cancer cellPC-3, MDA-MB-231 and DU145 ↓mTOR, ↑ activated AMPK/ULK1[132]
Baicalein and cisplatinGastric cancerSGC-7901 and SGC-7901/DDP↓Akt/mTOR and Nrf2/Keap 1[139]
BaicaleinThyroid cancerMDA-T68 ↑Bax, ↓NF-kB, ↓Cyclin B1[162]
BaicalinBladder cancer T24 cells↑Atg 5, ↑Atg 7, ↑Atg 12, ↑Beclin-1, ↑LC3-II [163]
BaicalinOsteosarcomaHOS, MG63 U2OS and 143B↓PI3K/Akt/mTOR, ↓ERK1/2, ↓β-catenin, ↑Bax, ↑caspase-3, ↑cleaved PARP[140]
BaicalinHepatocellular cancerSMMC-7721↓CD147, ↑Beclin-1[164]
ScutellarinBreast cancerMCF-7 ↑p-YAP, ↓Y.A.P., ↑autophagy[151]
ScutellarinLung cancerPC-9 and H1975, HepG2, Hela ↑LC3-II, ↓p-AKT[146]
Scutellarin and cisplatinLung adenocarcinomaA549, PC-9, H1975, and A549/DDP↑p53 and ↓c-met/AKT [165]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tuli, H.S.; Bhushan, S.; Kumar, A.; Aggarwal, P.; Sak, K.; Ramniwas, S.; Vashishth, K.; Behl, T.; Rana, R.; Haque, S.; et al. Autophagy Induction by Scutellaria Flavones in Cancer: Recent Advances. Pharmaceuticals 2023, 16, 302. https://doi.org/10.3390/ph16020302

AMA Style

Tuli HS, Bhushan S, Kumar A, Aggarwal P, Sak K, Ramniwas S, Vashishth K, Behl T, Rana R, Haque S, et al. Autophagy Induction by Scutellaria Flavones in Cancer: Recent Advances. Pharmaceuticals. 2023; 16(2):302. https://doi.org/10.3390/ph16020302

Chicago/Turabian Style

Tuli, Hardeep Singh, Sakshi Bhushan, Ajay Kumar, Poonam Aggarwal, Katrin Sak, Seema Ramniwas, Kanupriya Vashishth, Tapan Behl, Rashmi Rana, Shafiul Haque, and et al. 2023. "Autophagy Induction by Scutellaria Flavones in Cancer: Recent Advances" Pharmaceuticals 16, no. 2: 302. https://doi.org/10.3390/ph16020302

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop