Next Article in Journal
Exploring Nitric Oxide (NO)-Releasing Celecoxib Derivatives as Modulators of Radioresponse in Pheochromocytoma Cells
Next Article in Special Issue
Recent Advances in Natural Product-Based Hybrids as Anti-Cancer Agents
Previous Article in Journal
Synthesis and Biomedical Applications of Highly Porous Metal–Organic Frameworks
Previous Article in Special Issue
Structure Activity Relationship Studies around DB18, a Potent and Selective Inhibitor of CLK Kinases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chemistry and the Potential Antiviral, Anticancer, and Anti-Inflammatory Activities of Cardiotonic Steroids Derived from Toads †

1
International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
2
Pharmacognosy Group, Department of Pharmaceutical Biosciences, Biomedical Centre, Uppsala University, Box 591, SE 751 24 Uppsala, Sweden
3
International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing (Jiangsu Education Department), Zhenjiang 212013, China
4
Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom 32512, Egypt
5
Chemistry Department of Medicinal and Aromatic Plants, Research Institute of Medicinal and Aromatic Plants (RIMAP), Beni-Suef University, Beni-Suef 62514, Egypt
6
School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
7
Biochemistry Division, Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
8
Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, Okayama 7008530, Japan
9
Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
10
Zoology Department, Faculty of Science, Menoufia University, Shebin El-Kom 32512, Egypt
11
Pharmacognosy Department, College of Pharmacy, Tanta University, Elguish Street, Tanta 31527, Egypt
12
National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116024, China
13
Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
14
H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi 75270, Pakistan
15
Medical Malacology Department, Theodor Bilharz Research Institute, Imbaba, Giza 12411, Egypt
16
Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
17
Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE 10691 Stockholm, Sweden
*
Author to whom correspondence should be addressed.
Dedicated with great pleasure and honor to Lars Bohlin, Uppsala University, Sweden and Rob Verpoorte, Leiden University, Netherland on the occasion of their 75th birthday.
Molecules 2022, 27(19), 6586; https://doi.org/10.3390/molecules27196586
Submission received: 12 August 2022 / Revised: 30 September 2022 / Accepted: 30 September 2022 / Published: 5 October 2022
(This article belongs to the Special Issue New Anticancer Agents Based on Natural Products)

Abstract

:
Cardiotonic steroids (CTS) were first documented by ancient Egyptians more than 3000 years ago. Cardiotonic steroids are a group of steroid hormones that circulate in the blood of amphibians and toads and can also be extracted from natural products such as plants, herbs, and marines. It is well known that cardiotonic steroids reveal effects against congestive heart failure and atrial fibrillation; therefore, the term "cardiotonic" has been coined. Cardiotonic steroids are divided into two distinct groups: cardenolides (plant-derived) and bufadienolides (mainly of animal origin). Cardenolides have an unsaturated five-membered lactone ring attached to the steroid nucleus at position 17; bufadienolides have a doubly unsaturated six-membered lactone ring. Cancer is a leading cause of mortality in humans all over the world. In 2040, the global cancer load is expected to be 28.4 million cases, which would be a 47% increase from 2020. Moreover, viruses and inflammations also have a very nebative impact on human health and lead to mortality. In the current review, we focus on the chemistry, antiviral and anti-cancer activities of cardiotonic steroids from the naturally derived (toads) venom to combat these chronic devastating health problems. The databases of different research engines (Google Scholar, PubMed, Science Direct, and Sci-Finder) were screened using different combinations of the following terms: “cardiotonic steroids”, “anti-inflammatory”, “antiviral”, “anticancer”, “toad venom”, “bufadienolides”, and “poison chemical composition”. Various cardiotonic steroids were isolated from diverse toad species and exhibited superior anti-inflammatory, anticancer, and antiviral activities in in vivo and in vitro models such as marinobufagenin, gammabufotalin, resibufogenin, and bufalin. These steroids are especially difficult to identify. However, several compounds and their bioactivities were identified by using different molecular and biotechnological techniques. Biotechnology is a new tool to fully or partially generate upscaled quantities of natural products, which are otherwise only available at trace amounts in organisms.

1. Introduction

Cardiotonic steroids (CTS) were documented by ancient Egyptians more than 3000 years ago [1]. It is well known that cardiotonic steroids show therapeutic activity against congestive heart failure; therefore, the term "cardiotonic" has been coined. Cardenolides can also be used to treat atrial fibrillation by increasing intracellular Na+ and controlling the contraction of cardiac fibers [2]. The sodium pump is composed of two subunits in equimolar ratios: (i) the α-catalytic subunit which is a multipass transmembrane protein containing the binding sites for Na+, K+, ATP, and CTS, and (ii) the β regulatory subunit, a transmembrane protein with several glycosylation sites [3]. The positive inotropic activity of cardiotonic steroids that mediate clinically useful physiological effects in patients has been attributed largely to high-affinity inhibitory interaction with the extracellular surface of the membrane-bound sodium pump (Na+/K+-ATPase) [4]. However, CTS were first reported to be vital in the regulation of renal sodium transport and arterial pressure. Recent epidemiological evaluations have implicated that these types of steroids have an intrinsic role in the regulation of cell growth, differentiation, apoptosis, and fibrosis, the modulation of immunity, and the control of various central nervous functions and even behavior [5]. According to Dvela et al., compounds that are derivatives from CTS have been used for centuries to treat cardiac failure and arrhythmias in Western and Eastern medicine [6]. CTS also contribute to pro-hypertrophic and pro-fibrotic cell signaling [7]. CTS are divided into two distinct groups, i.e., cardenolides, such as ouabain and digoxin, and bufadienolides, such as marinobufagenin, telocinobufagin, and bufalin [8], as shown in Figure 1. In general, herbs and animal preparations containing CTS have been used for centuries as emetics, diuretics, and arrow poisons [9].
Bufadienolides are one type of CTS comprising two substances, bufalin (BF) and marinobufagenin. They were first primarily isolated from the skin of Bufo toads (formerly Bufo marinus, now Rhinella marina L.) and were used to treat heart diseases in traditional Chinese medicine [1]. The use of digitalis, ouabain, and strophanthin glycosides to slow the rate and strengthen the contractility of the heart is one of the most important strategies to combat cardiac failure. Among these agents, digitalis glycosides are the most widely used compounds. Their positive inotropic and negative chronotropic effects on cardiomyocytes are related to the coupling between Na+/K+ ATPase and Na+/Ca2+ exchanger through their intracellular concentration.
There are extraordinary arsenals of chemicals in the animal kingdom that are critical for their survival, defending against predators and hunting [10,11]. The mucous glands are a significant source of bioactive compounds due to their evolutionary adaptations [12,13]. The skin of toads is distinguished by serous and mucous glands which release a variety of chemicals that aid in the defense against poisoning predators [13,14]. Mucous glands are many and slightly smaller. They are crucial for maintaining the skin’s pH, moisturizing it, and secreting a cocktail of bioactive compounds [15]. In addition, cardiotonic steroids have a wide spectrum of pharmacological activities, among them cardiotonic, anti-arrhythmic, antidiabetic, immunomodulatory, antibacterial, antifungal, antiprotozoal, antiviral, antineoplastic, sleep inducing, analgesic, contraceptive, endocrine activity, behavioral changes, and wound healing [16]. Clinically, CTS have demonstrated potent inhibition of cardiac Na+ and K+-ATPase (NKA), the integral membrane protein that maintains ionic gradients in cells, and therefore, they have potential activity in heart failure treatment [17]. This interaction inhibits the enzyme, stops the transport of sodium to the outside of the cell and potassium to the inside of the cell, and activates the calcium pump, which increases cardiac contraction [18]. The serous granular glands are intimately associated with the defense mechanisms against predators. These glands are scattered across the skin’s surface or clustered next to each other, similar to parotoid glands. Although the genus Bufo has parotoid glands mostly on the head, neck, or shoulder [19], the parotoid glands are more often seen as paired protuberances in the postorbital place in the Anura [15,20]. Targeting the Na+/K+ ATPase pump has been a focus of numerous research groups to reveal the pharmacological impacts of CTS, including cardiotonic, anticancer, anti-inflammatory, and antiviral activities [21,22,23,24].

1.1. Synthesis and Factors Affecting Poison Composition

Toads employ a complex series of protective processes, beginning with expanding the lung, elevating the body, and spraying the venom. External pressure is likely the most critical factor in the release of parotoid venom. Venom jets are not released from the parotoids until adequate mechanical pressure is applied. Additionally, lung inflation contributes to the expansion of lung pressure, which may be moved over to the parotoid floor and passed to the bottle-shaped parotoid glands. Consequently, the secretion is forced out via the duct slit, enabling venom jets to spray out [20].
While food has a role in the formulation of toad poison, bufadienolides can be produced through the metabolism of bacteria that colonize the parotoid glands. Bufadienolides can undergo various structural transformations, resulting in a high degree of structural diversity in this family of metabolites [25]. Early in larval development, bufadienolides are produced [26]. Toads of common occurrence Bufo bufo L. produce toxic or unappealing bufadienolides for various predators [27]. Bufadienolides have been reported to be more diverse and abundant in toad larvae in natural populations with a higher competitor density [28] and correspondingly increased when tadpoles have been subjected to food limitations in the laboratory [26].
If threatened, Rhinella jimi Stevaux typically shows a stereotyped defensive behavior, which includes inflating the lungs and generating a stiffened and voluminous posture, usually accompanied by a head-butting pattern, wherein the animal tilts its body in the direction of the dangerous and potentially agent, exposing one of the parotoids [13]. Head-butting appears to be a major component of a toad’s defense strategy, since exposing the parotoid improves the likelihood of the animal’s venom being triggered and delivered before the predator can bite it.

1.2. Biosynthesis of CTS of Toad Origin (Bufadienolides)

Despite substantial progress in the structure elucidation and mechanisms of the action of CTS (bufadienolides), the biosynthesis of these endogenous CTS is still poorly understood. Since the enzymes responsible for the synthesis of specific bufadienolides have not yet been identified, the ability to develop knockout and/or overexpression models is also limited so far. Chiadao and Osuch (1969) evaluated bufadienolide biosynthesis via injection of some precursors labeled with 14C or tritium into toads (Bufo marinus), which included Cholesterol-4-14C, pregnenolone-4-14C, methyl 3β-formoxy-5β-cholanate-24-14C, sodium 3β-hydroxy-5β-cholanate-24-14C, methyl 3β-acetoxy-5-cholenate-3H, methyl 3β-hydroxy-5α-cholanate-3H, and methyl 3α-hydroxy-5β-cholanate-24-14C. It was found that both 5α and 5β, 3β-hydroxycholanate derivates were more competent precursors for biosynthesis, as shown in Figure 2 [29]. However, according to Siperstein et al., cholesterol is the precursor responsible for the synthesis of marinobufagin by the B. marinus toad [30] (Figure 3). Moreover, [2-14C] mevalonic acid, [20-14C] pregnenolone, and [20-14C] cholesterol were evaluated as precursors for bufadienolides biosynthesis in Bufo pnracnemis toads by Proto et al. It has also been reported that both pregnenolone and mevalonic acid were poor precursors in winter (natural hibernation period), but mevalonic acid was involved in the summer (active life of the toad), while cholesterol was potent in both seasons of the year. The outcomes also indicated that the double unsaturated δ-lactone ring of the bufadienolides was directly derived from the cholesterol side chain without the compound’s prior conversion into pregnenolone [31].

1.3. Structure and Morphology of Glands Secreting Venom in Toad

Poison glands have been generated in anuran amphibians by a singular multinucleated cytoplasm mass that produced a secretory syncytium. Lumen did not exist, and a matrix of syncytial cytoplasm surrounded the gland. After being generated in the syncytial periphery, the granular secretions were maintained across the cytoplasm [32]. Individuals or clusters of glands can be seen in certain parts of the body. They resemble warts, which are frequently observed on the dorsal epidermis of toads (bufonids) if gathered [13]. If they occur in massive quantities, they can produce prominent protrusions called macro-glands [13,33], amongst which even the parotoids are unquestionably the most prevalent. Due to the huge size of the parotoid individual glands, these cutaneous systems can produce and store considerable quantities of poison for chemical defense against predators [20,34]. Parotoids in the Rhinella genus toads are histologically described as huge stockpiles of gigantic poison glands implanted into the dermis and grouped side by side to form honeycomb-like structures [35,36,37]. Salamanders [38], phyllomedusin-producing tree frogs [36], frogs [34], and toads [20] all have parotoids. They have a duct that is surrounded by a layer of myoepithelial cells. As compared with skin poison glands, the epithelial that coats the duct internally is extremely dense, obstructing the ductal canal and occasionally leaving only a tiny split in the middle [20,36,38,39]. Distinguishable mucous glands are also referred to as accessory glands, because they encircle each other [20,36,39]. Two bufadienolides have been isolated from the secretion of Rhinella jimi parotoid macro-glands, and both showed action against Leishmania chagasi Nicolle [40,41,42].

2. Chemistry of Cardiotonic Steroids (CTS)

The chemistry of steroids has been investigated thoroughly, including steroid glycosides isolated from toads, sponges, star fishes, fungi, and echinoderms [43,44,45,46]. However, toads cardiotonic steroids are specifically difficult to identify, and therefore, are less explored as compared with their terrestrial analogues.
The term CTS is commonly use as a synonym for cardiac glycosides (CGs), owing to their commonly glycosylated steroid nuclei and their potential biological activity. They are a unique category of phytosteroids that can be classified into two types based on their source and chemical feature: (1) cardenolides and (2) bufadienolides (Figure 1), isolated from the skin and the parotoid gland of toads [47].
Structure activity relationships (SAR) of CGs have been studied based on natural and semisynthetic analogues, their abilities to bind with sodium pump receptors, i.e., Na+/K+ ATPase pump, and subsequent cardiotonic effects. The SARs were summarized by Melero et al. and Gopalakrishnakone et al. [21,48]. The understanding of SAR has helped the synthesis of semisynthetic cardenolides with more potency than the native molecules [49].
Cardenolides (C23) and bufadienolides (C24) both bear common structural features such as the cis-fused C/D rings with a hydroxy group positioned at 14β. The major difference between them involves the lactone ring at the 17β position. Hence, the basic sterane (cyclopentanoperhydrophenanthrene) structure in this unique cis-trans-cis-configuration makes the CTS distinct from other steroidal compounds such as bile acids, sterol, or steroid hormones [50].
Cardenolides possess an unsaturated lactone ring substituent, i.e., α, β-unsaturated five-membered butyrolactone ring (but-2-en-4-olide ring), while bufadienolides have a six-membered unsaturated α-pyrone ring (penta-2,4-dien-5-olide ring) [47,51], since saturation of the lactone ring attenuates the CT bioactivity [50]. In addition, some CTS are usually found as sugars, where the aglycone part (cardiotonic steroid) is linked at the 3β-OH group to mono- or oligosaccharide sugar chains (maybe one, two, three, or four sugars) named glycone or sugar moiety. The sugars are thought to be D-glucose, L-rhamnose, L-arabinose, and D-xylose (Figure 1) [52,53]. In general, there are homo-oligomer and hetero-oligomer CTS. For example, scillaren A is a hetero-oligomer bufadienolide, which is glycone-structured rhamnose/glucose, while digoxin is a homo-oligomers, which is glycone composed of three units of digitoxose [51]. The most common glycosidic bonds connecting saccharide units are O-glycosidic bonds in which the oxygen from a hydroxyl group becomes linked to the carbon atom [54]. This glycone moiety enhances the water solubility of the glycoside and its ability to bind to its molecular target, i.e., Na+/K+-ATPase or the sodium pump in the heart muscle [50,55]. Hence, there are many sites for interaction via hydrophobic binding by steroidal nucleus and sugar moiety, in addition to the electrostatic and hydrogen bonding by the lactone ring [48,56].
Nevertheless, terrestrial plants and animal CTS are still underestimated and, consequently, less medicinally explored. The well-known CGs include those isolated from frog venoms. These compounds are mainly bufadienolides.
Toads’ (34 genera and 410 species [57]) CTS are of the bufadienolide type and occur either as free genins (bufagins) or glycosides bound to suberylarginine (bufotoxins) [48].
These toxins are found in the skin of parotoid glands and, consequently, can be extracted from dried secretion of the auricular and skin glands of toads. With the aid of metabolomics, specifically with metabolite profiling with ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) of toad secretions, 31 bufadienolide metabolites have been identified [58]. Some examples of the CTS are shown in Figure 4. The major differences among these compounds are the hydroxylation at 11α either alone as in gamabufagin isolated from Bufo japonicus Temminck & Schlegel and 11α-hydroxytelocinobufagin in Rhinella marina L. or at 19β, as in 11α, 19-dihydroxy-telocinobufagin in R. marina. Other structural modifications may also involve 14β, 15β-epoxy ring formations as in marinobufagenin and resibufogenin found in Bufo rubescens (now Rhinella rubescens Lutz) and Bufo gargarizans Cantor, respectively [58].
Based on Kamano et al. and Gopalakrishnakone et al., structural modifications in the BF molecule can increase or decrease the cytotoxic activity against primary liver carcinoma cells PLC/PRF/5. For example, the acetylation of the 3β-hydroxyl group, 11α-hydroxyl substituent, and aldehyde group at 19 have been shown to increase the antitumor effect, while 5β-hydroxyl, 16β-acetoxy, and 19-CH2-OH substituents decreased the activity [21,59]. In addition, microbial biotransformation of cinobufagin has been reported by the filamentous fungus Alternaria alternata (Fr.) Keissl. (Fr.), explained by the production of 12β-hydroxyl and 3-oxo-12α-hydroxyl derivatives. The products exhibited decreased but still significant in vitro cytotoxic activities [60].

3. Antiviral Activity

In particular, the antiviral effects have recently attracted interest as compared with the well-documented antitumor activity [61,62,63,64]. Inhibition of the transmembrane Na+/K+ ATPase protein affects cell host intracellular signaling processes and viral life at different levels, resulting in inhibition of the viral genome expression, replication, protein translation, release, and entry. Nevertheless, these activities have been thoroughly investigated for CTS [65,66,67,68].
The CTS with antiviral activity are reported in Table 1. BF is active against coronaviruses, i.e., severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Middle East respiratory syndrome coronavirus (MERS-CoV) via inhibition of Src signaling mediated by ATP1A1 and virus entry into the host cells [66,69]. In a comparative study among CTS, bufalin, cinobufagin, and telocinobufagin had high anti-MERS-CoV activity, and bufalin had the most potent anti-SARS-CoV and SARS-CoV-2 activity [70]. The most potent anti-coronaviral compounds were, cinobufagin, telocinobufagin, followed by bufalin. They acted by downregulating the cell death-related genes as well as the immune- and inflammatory-related genes to balance the levels of C/D-class small nucleolar RNAs (SNORDs) and H/ACA small nucleolar RNAs (SNORAs). Additionally, the toxicities of bufalin, cinobufagin, telocinobufagin, bufotalin, and cinobufotalin were tested in vivo by Jin et al., 2021, suggesting that bufalin had the highest anti-coronaviral activity as well as the strongest toxicity. Thus, cinobufagin and telocinobufagin were selected for their high anti-coronavirus activity and low toxicity [70]. Moreover, cinobufacini (BF and cinobufagin) has been reported to inhibit mRNA expression of the hepatitis B virus (HBV) [71]. BF (15 nM) also has a higher potency than ouabain and digoxin as an anti-HSV to reduce viral yield by 90% [68]. Cinobufagin and resibufogenin showed 50% inhibitory concentrations (IC50) against enterovirus 71 (EV71) infection in vitro at 10.9 ± 2.4 and 218 ± 31 nM, respectively. EV71 is a pathogen affecting hand, foot, and mouth disease (HFMD) that induces CNS inflammation and life-threatening systemic complications, i.e., cardiorespiratory failure. Viral protein synthesis are likely to be targeted by both compounds [72]. BF and cinobufagin also inhibit HIV-1 with an IC90 at 15 and 40 nM, respectively [73].

4. Anticancer Activity

The anticancer activities of cardiotonic steroids (CTS) from different toad species are shown in Table 2. Cardiotonic steroids or cardiotonic glycosides are characterized by their abundance in nature, diversity of structure, potential for chemical modification, and wide use in heart failure management. CTS act by binding to the extracellular surface of Na+/K+-ATPase. The altered expression of the sodium pump subunits in different cancers strongly suggests that targeting Na+/K+-ATPase represents a novel means to fight the growing number of cancers [74]. Cardiotonic action occurs via inhibition of Na/K-ATPase, mediating cardiac muscle contraction [75]. Inhibition of Na/K-ATPase increases the influx of Na+ followed by a decrease of K+ efflux in heart cells. Increases in Ca2+ ions raise the cardiac contractile force, since more Ca2+ is available for the involved proteins. CGs trigger the accumulation of intracellular Ca2+ levels leading to an increase in muscle tone and the circulating blood volume per minute, leading to the control of heart rate and stroke volume [76]. Numerous studies have evaluated changes in the transmembrane transport of cations during the course of malignant cell transformation, due to an increase in Na+/K+-ATPase activity [77,78,79,80]. There is evidence that these kinetic changes in Na+/K+-ATPase activity are already present at very early stages of tumorigenesis, even before the morphological alteration and the abnormal appearance of tumors [81,82]. CTS stimulate protein tyrosine phosphorylation and a number of growth-related pathways in a cell- and tissue type-dependent manner [83,84,85,86,87,88]. CTS exhibit in vitro cytotoxic and cytostatic effects against various human cancer cell lines, which is attributable to their ability to induce cell-type-specific cell death modalities [89]. Additionally, selected CGs have entered phase I and II of clinical trials for the treatment of solid tumors with satisfactory safety and efficacy [90,91].
The antiproliferative activity of CGs on different types of cancer cell lines, including those of breast cancer, prostate cancer, pancreatic cancer, leukemia, neuroblastoma, and melanoma, have been previously described [86,92]. The predominant theory for the mode of action of CGs in tumor cells focuses on their binding to the α-subunit of Na/K-ATPase, leading to changes in pumping activity, which increases the intracellular Na+ levels and depletes the K+ levels. Consequently, the intracytoplasmic Ca2+ levels increase owing to exacerbation of mitochondrial Na+/Ca2+ exchange. Other theories have suggested that CGs may interact with the plasma membrane via the steroid nucleus, causing a change in membrane fluidity and indirectly affecting the function of several membrane proteins and receptors [93,94]. The sodium pump is composed of multifunctional groups of α and β subunits. The α-1 subunit of the sodium pump is overexpressed in some types of cancer, including non-small cell lung cancer (NSCLC), renal carcinoma, glioma, and melanoma, whereas the alfa-3 subunit is overexpressed in colon carcinoma [93,95,96]. In addition, in mice, α-2, α-3, and α-4 isoforms are naturally susceptible to CGs. In rodents, the α-1 isoform is resistant to the binding of ouabain and, in humans, the α -1 isoform is sensitive to CGs and may, therefore, play a key role in the signal transduction pathways [97].
CGs can induce several signal transduction pathways such as inhibition of DNA topoisomerase II [98]; inhibition of the nuclear factor-κ light-chain-enhancer of activated B cell (NF-κB)-mediated pathways; changes in cell cycle, specifically blocking S and G2/M phases; inhibition of interleukin (IL)-8 production [99]; and modulation of the myeloid cell leukemia-1 (Mcl-1) as an essential factor for cell death [5,100]. CGs and particularly ouabain are involved in metastatic cascades by inhibiting the migration of H292 lung tumor cells via the suppression of regulatory migration proteins, such as focal adhesion kinase (FAK) and Akt [101]. Furthermore, digitoxin has been shown to inhibit angiogenesis in human umbilical vein endothelial cells (HUVEC) and promoted FAK activation by several pro-angiogenic stimuli [102]. Gamabufotalin has been shown to inhibit vascular endothelial growth factor (VEGF)-triggered HUVEC proliferation, migration, and invasion in vitro by suppressing the VEGF receptor-2 signaling pathway [103].
BF-induced apoptosis in human leukemia cells [104] is mediated by ERK-kinase cascade which is excessively activated in order for BF-mediated apoptosis to occur [105,106,107]. Treatment of human leukemia THP-1 cells with BF-induced inflammatory cytokine interleukin-1 beta (IL-1β) and tumor necrosis factor-α (TNF-α). After treating the cells with an inhibitor of ERK, i.e., PD-98059, the cytokine production was attenuated, suggesting that the ERK pathway was responsible for the inflammatory response induced by BF [105].
BF exerts an antitumor effect by inducing apoptosis and triggering autophagic cell death in various human cancer cells. The anti-inflammatory activities of BF are also important for its antitumor function [107]. BF has been shown to inhibit proliferation and induced mitochondria-dependent apoptosis in U2OS and Saos-2 cells [108]. Furthermore, a mechanistic investigation demonstrated that BF was able to significantly decrease Mcl-1 expression level and modestly decrease Bcl-XL expression level. The downregulation of these anti-apoptotic proteins has been well correlated with deactivation of transcription factor STAT3. BF and cinobufagin (1–10 μM) inhibited the proliferation and induced cell apoptosis of androgen-dependent (LNCaP) and independent (DU145 and PC3) prostate cancer cell lines [109]. Qi et al. [110] found that BF and cinobufagin induced apoptosis and increased the proportion of apoptotic cells. This apoptotic induction was associated with an increase in Fas, Bax, and Bid expression; a decrease in Bcl-2 expression; disruption of the mitochondrial membrane potential; release of cytochrome c; activation of caspase-3, -8, -9, and -10; and the cleavage of poly (ADP-ribose) polymerase (PARP), which indicated that BF and cinobufagin induced apoptosis through both Fas and mitochondria-mediated pathways [111]. BF also inhibited APL cell proliferation in a time- and dose-dependent manner and induced NB4 cell apoptosis accompanied by downregulation of survivin and activation of caspase-3. The MEK/ERK signaling pathway was negatively regulated in BF-induced apoptosis in the NB4 human leukemia cell line. BF enhanced ATRA-induced differentiation in NB4 cell line and primary culture in APL cells [112], and also induced MGc-803 cell death characterized by apoptotic phenotype DNA content changes and chromosome DNA fragmentation. BF triggered cell cycle arrest in G1 phase, leading to the induction of apoptosis of gastric cancer cells [113]. A distinct study has concluded that BF-induced apoptosis by altering the expression of apoptosis-related genes c-Myc and Bcl-2 [114], and the activation of mitogen-activated protein kinase (MAPK) may be involved in BF-induced apoptosis in U93T cells [115]. Overexpression of Bcl-2 inhibited BF-induced MAPK activation and the subsequent AP-I activation and cell apoptosis in U937 cells [113].
BF was able to suppress the migration and invasion of prostate cancer cells through HOTAIR, the sponge of miR-520b [115]. In addition, the same natural product inhibited human breast cancer tumorigenesis by inducing cell death through the ROS-mediated RIP1/RIP3/PARP-1 pathways [116]. BF also attenuated the proliferation of breast cancer MCF-7 cells in vitro and in vivo, by inhibiting the PI3K/Akt pathway, and the proliferation of breast cancer MCF-7 cells in vitro and in vivo by inhibiting the PI3K/Akt pathway [117]. The toad skin extract cinobufatini inhibited migration of human breast carcinoma MDA-MB-231 cells in a model stromal tissue [118], while key members of bufadienolides, i.e., BF, bufotalin, and gamabufotalin, have significantly sensitized human breast cancer cells with different status of ER-α to apoptosis induction of TRAIL, as evidenced by enhanced Annexin V/FITC positive cells (apoptotic cells), cytoplasmic histone-associated DNA fragments, membrane permeability transition (MPT), caspases activation, and PARP cleavage [119]. A BF derivative has exhibited stronger apoptosis-inducing effect than BF in A549 lung cancer cells and lower acute toxicity in mice [120]. The original natural product induced apoptosis of lung cancer cells via the regulation of the PI3K/Akt pathway. [121] used the A549 human lung adenocarcinoma epithelial cell line as an experimental model to evaluate the effects of BF in lung cancer chemotherapy. BF suppressed colorectal cancer cell growth through promoting autophagy in vivo and in vitro [122]. In addition, it was able to inhibit hTERT expression and colorectal cancer cell growth by targeting cpsf4 [123], to reverse ABCB1-mediated drug resistance in colorectal cancer [124], and to regulate mTOR and ERK signaling pathways in gastric cancer cells [125]. BF treatment could decrease miR-298 expression. Previously, we have also shown that the deletion of miR-298 contributed to BF-induced apoptosis in gastric cancer cells by targeting BAX, an apoptosis protein [126]. BF revealed anticancer effects on human hepatocellular carcinoma HepG2 cells [127].
The bufadienolides are a group of steroid compounds belonging to CGs, a class of circulating substances [128,129,130,131]. Marinobufagenin (MBG), an endogenous mammalian cardiotonic agent [132,133], is currently the most commonly used in drug research. MBG has been implicated in various physiological conditions [134] and appeared to be associated with pathophysiological events in animals [135,136,137] and humans [138,139,140]. MBG inhibits glioma growth in vivo and in vitro through the sodium pump α1 subunit and ERK signaling-mediated mitochondrial apoptotic pathways, and activates ERK/NF-κB signaling, activating the caspase signaling cascade through accelerating the release of cytochrome C, inducing apoptosis. These data support the potential use of MBG as an anticancer agent [141].
Resibufogenin (RB) is a commonly used natural medicinal compound extracted from Asiatic toad Bufo gargarizans Cantor. This compound is known for its analgesic, anti-inflammatory, anticancer, and anti-radiation properties, as well as its cardio protective and anesthetic activities. RB effectively inhibits cell viability, and induces apoptosis, caspase-3, and caspase-8 activities in MGC-803 cells by suppressing the PI3K/AKT/GSK3β signaling pathway. Therefore, it may be considered as a possible treatment for gastric carcinoma [142]. RB suppresses ovarian cancer growth and glycolysis in vitro and in vivo. Moreover, the downregulation of PIM1 by RB plays a key role in the anticancer activities of RB [143]. RB suppresses colorectal cancer growth and metastasis, activating RIP3 and phosphorylating MLKL, leading to necroptosis [144]. RB suppresses transforming growth factor-β-activated kinase 1-mediated nuclear factor-κB activity through protein kinase C-dependent inhibition of glycogen synthase kinase 3 [145].

5. Anti-Inflammatory Activity

Some research has suggested that cardiotonic steroids, including bufalin, have immunomodulatory properties [153], as they interact with numerous inflammatory responses such as vascular permeability, cell migration, and proinflammatory cytokines [153,154]. The main CTS with anti-inflammatory activity are shown in Table 3. Carvalho et al. [155] demonstrated that the anti-inflammatory activity of obufagenin in vivo and in vitro exposed a novel endogenous function for this steroid in mammals, owing to its ability to reduce cytokine levels in peritoneal macrophage culture (Table 2).
Bufalin has a long history of being used as an anti-inflammatory medicinal drug in China and other Asian countries [156]. Wen et al. [157] investigated bufalin’s analgesic and anti-inflammatory properties in vivo, suggesting that it could be a possible drug treatment for inflammatory diseases. They observed that bufalin effectively suppressed NF-κB activation in vivo by preserving IκBα levels, decreasing the nuclear translocation of NF-κB p65, and inhibiting downstream proinflammatory mediators. Ye et al. [158] demonstrated that bufalin suppressed the nuclear translocation of NF-κB in response to TNF in vitro. In addition, bufalin has been shown to control NF-κB activity [159], a key regulator of the inflammatory process that plays a significant role in inflammation. It modulates the expression of proinflammatory mediators, including cyclooxygenase-2, IL-6, inducible nitric oxide synthase, and TNF, IL-1β [160]. NF-κB signaling is the ideal therapeutic target for inflammation pathogenesis. Furthermore, the bufalin substitutes, ouabain and digoxin, have potent anti-inflammatory properties [161,162]. Ye et al. [158] revealed that bufalin suppressed tumor necrosis factor (TNF) signaling in human 293T cells via interfering with NF-κB nuclear translocation. Zhakeer et al. [9] demonstrated that bufalin attenuated hyperresponsiveness and suppressed increases of total inflammatory cells in a mouse asthma model. The levels of IL-4, IL-5, and IL-13 in serum were significantly reduced. Cell infiltration, goblet cell hyperplasia, IκBα degradation from NF-κB, and the level of phosphorylated p65 protein levels in the lung tissues were all decreased, indicating that it may mediate its anti-inflammatory effects via inhibiting NF-κB activity. Bufalin may inhibit the activation of NF-κB and may reduce the production of its downstream proinflammatory mediators during acute inflammation [157].

6. CTS with Antiviral, Anticancer, and Anti-Inflammatory Properties

The potential pharmaceutical properties of bufadienolides isolated from toad venom have been investigated recently. Anticancer activity of bufalin has been seen against breast, liver, and gastric and leukemia cancer cells, as listed in Table 1. It has been indicated that bufalin inhibited tumor growth through apoptosis induction by multiple pathways [165]. In in vivo studies, bufalin suppressed human hepatocellular carcinoma (HHC) cell growth and induced apoptosis by activating Bax without noticeable toxicity [114]. In another study, nude mice injected with HCCLM3-R cells were studied after bufalin treatment. Significant antitumor activities, manifested with the regress noticed in the metastatic growth in parallel with the inhibition of AKT/GSK3/catenin/E-cadherin signaling pathways, were evident [166]. Bufalin inhibited tumor growth by inducing cell apoptosis through the intrinsic apoptotic pathway. A reduction in tumor size of the human lung cancer cell line, NCI-H460, was confirmed after bufalin treatment without significant drug-related toxicity [167].
Anti-inflammatory and anticancer effects of bufalin have been illustrated through the inhibiting NF-B pathway, which is an important pathway in both anti-inflammation and cancer [168,169]. Bufalin reduces hyperresponsiveness, and inhibits the OVA-induced activation of inflammatory cells, including macrophages, eosinophils, lymphocytes, and neutrophils, as well as cytokines, including IL-4, IL-5, and IL-13. Additionally, a reduction in inflammatory cell infiltration and goblet cell hyperplasia and blockage of NF-B have been noticed [9]. The anti-inflammatory and analgesic effects of bufalin have been studied in a carrageenan-induced paw oedema model. Bufalin downregulated the expression of nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin-1 (IL-1), interleukin-6 (IL-6), and tumor necrosis factor (TNF), to which the inhibitory effect on the master switch of NF-B signaling was attributed [157]. Moreover, bufalin had high anti-MERS-CoV activity, and had the most potent anti-SARS-CoV and SARS-CoV-2 activity [70].

7. Conclusions

Using natural products as therapeutic agents has gained much attention, therefore, it is noteworthy to document the recent updates in the literature with particular emphasis on the possible applications of cardiotonic steroids in the promotion of anticancer and antiviral strategies. Limited numbers of CTS have been expected to have anticancer activity. Cardenolides, mainly attributed to digitalis and digoxin, have potential effects in the treatment of congestive heart failure and atrial fibrillation by binding to the extracellular membrane and activating Na+/K+-ATPase. Targeting Na+/K+-ATPase and altering the expression of the sodium pump subunits represents a novel means to fight the growing number of cancers. For instance, marinobufagenin has been shown to inhibit the growth of glioma cancer in vivo and in vitro via the sodium pump α1 subunit and ERK signaling-mediated mitochondrial apoptotic pathways. Additionally, γ bufotalin has been reported to suppress vascular endothelial growth factor (VEGF)-triggered proliferation, migration, and invasion by suppressing the VEGF receptor-2 signaling pathway in vitro. Similarly, resibufogenin (RB) is a known anticancer agent that acts via the inhibition of cell viability, induces apoptosis, caspase 3, and caspase 8 activities in MGC 803 cells by suppressing the PI3K/AKT/GSK3β signaling pathway. Finally, bufalin (BF) is active against corona viruses, i.e., SARS-CoV-2 and MERS-CoV, via inhibition of Src signaling mediated by ATP1A1, and thus, blocking entry of the virus into the host cells. As well, BF and cinobufagin inhibit mRNA expression of the hepatitis B virus (HBV) and HIV-1. Furthermore, cinobufagin and resibufogenin have been shown to prevent 71 (EV71) infections from enterovirous. In the current review, we illustrate the significance of cardiotonic steroids as potential anticancer, anti-inflammatory, and antiviral candidates.
Further studies are suggested to screen the efficacy of CTS isolated from toad species from different geographical regions, and under different environmental conditions to ensure stability of their chemical composition. Additional molecular mechanisms and modeling, drug safety, and quality control are needed to motivate the future implications of CTS as novel therapeutic agents that will certainly contribute to the development of pharmaceutical industry.

Author Contributions

Writing—original draft preparation, H.R.E.-S., B.E.-A., M.R.H., A.Z., S.A.M.K., N.Y. and S.H.M.; Interpretation of the data and visualization, M.M.H., H.E.T. and X.Z.; Revising and reviewing, Z.G., H.R.E.-S. and T.E.; Writing—review and editing, S.A.M.K., M.D., A.S., S.G.M., I.M.E.-G., N.Y., S.H.M. and H.R.E.-S.; Idea and project administration, S.A.M.K. and H.R.E.-S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

No applicable.

Informed Consent Statement

No applicable.

Data Availability Statement

No applicable.

Acknowledgments

El-Seedi, H.R thanks Jiangsu University for Adjunct Professor fellowship.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviation

Bufalin, BF; cardiac glycosides, CGs; CTS, cardiotonic steroids; cyclooxygenase-2, COX-2; cytomegalovirus, CMV; Chikungunya virus, CHIKV; epithelial–mesenchymal transition, EMT; enterovirus 71, EV71; extracellular regulated kinase, ERK; focal adhesion kinase, FAK; glycogen synthase kinase-3, GSK3β; human hepatocellular carcinoma, HepG2; human lung cancer cell line, NCI-H460; human T-cell leukemia virus type-1, HTLV-1; human immunodeficiency virus type-1, HIV-1; hepatitis B virus, HBV; human umbilical vein endothelial cells, HUVEC; herpes simplex virus, nHSV; IκB kinase, IKKβ; induced inflammatory cytokines interleukin-1 beta, IL-1β; lung cancer, A549; middle east respiratory syndrome coronavirus: MERS-CoV; myeloid cell leukemia-1, Mcl-1; membrane permeability transition, MPT; marinobufagenin, MBG; Middle East respiratory syndrome coronavirus, MERS-CoV; non-small cell lung cancer, NSCLC; nuclear factor kappa B, NFκB; nitric oxide synthase, iNOS; pancreatic carcinoma, PANC-1; poly ADP-ribose polymerase, PARP; phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling pathways, PI3K/Akt/mTOR pathway; phosphoinositide 3-kinases, PI3K; resibufogenin, RB; respiratory syncytial virus, RSV; structure activity relationships, SAR; severe acute respiratory syndrome coronavirus 2, SARS-CoV-2; signal transducer and activator of transcription 3, STAT3; tumor necrosis factor-α, TNF-α; vascular endothelial growth factor, VEGF.

References

  1. El-Mallakh, R.S.; Brar, K.S.; Yeruva, R.R. Cardiac glycosides in human physiology and disease: Update for entomologists. Insects 2019, 10, 102. [Google Scholar] [CrossRef] [Green Version]
  2. El-Bakry, A.A.; Hammad, I.A.; Galal, T.M.; Ghazi, S.M.; Rafat, F.A. Polymorphism in Calotropis procera: Variation of metabolites in populations from different phytogeographical regions of Egypt. Rend. Lincei 2014, 25, 461–469. [Google Scholar] [CrossRef]
  3. Mijatovic, T.; Van Quaquebeke, E.; Delest, B.; Debeir, O.; Darro, F.; Kiss, R. Cardiotonic steroids on the road to anti-cancer therapy. Biochim. Biophys. Acta Rev. Cancer 2007, 1776, 32–57. [Google Scholar] [CrossRef]
  4. Heasley, B. Chemical Synthesis of the Cardiotonic Steroid Glycosides and Related Natural Products. Chem.-A Eur. J. 2012, 18, 3092–3120. [Google Scholar] [CrossRef] [PubMed]
  5. Bagrov, A.Y.; Shapiro, J.I.; Fedorova, O.V. Endogenous cardiotonic steroids: Physiology, pharmacology, and novel therapeutic targets. Pharmacol. Rev. 2009, 61, 9–38. [Google Scholar] [CrossRef] [PubMed]
  6. Dvela, M.; Rosen, H.; Feldmann, T.; Nesher, M.; Lichtstein, D. Diverse biological responses to different cardiotonic steroids. Pathophysiology 2007, 14, 159–166. [Google Scholar] [CrossRef]
  7. Kolmakova, E.V.; Haller, S.T.; Kennedy, D.J.; Isachkina, A.N.; Budny, G.V.; Frolova, E.V.; Piecha, G.; Nikitina, E.R.; Malhotra, D.; Fedorova, O.V.; et al. Endogenous cardiotonic steroids in chronic renal failure. Nephrol. Dial. Transplant. 2011, 26, 2912–2919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Pavlovic, D. The Role of Cardiotonic Steroids in the Pathogenesis of Cardiomyopathy in Chronic Kidney Disease. Nephron Clin. Pract. 2014, 128, 11–21. [Google Scholar] [CrossRef] [PubMed]
  9. Zhakeer, Z.; Hadeer, M.; Tuerxun, Z.; Tuerxun, K. Bufalin Inhibits the Inflammatory Effects in Asthmatic Mice through the Suppression of Nuclear Factor-Kappa B Activity. Pharmacology 2017, 99, 179–187. [Google Scholar] [CrossRef]
  10. Abd El-wahed, A.; Yosri, N.; Sakr, H.H.; Du, M.; Algethami, A.F.M.; Zhao, C.; Abdelazeem, A.H.; Tahir, H.E.; Masry, S.H.D.; Abdel-daim, M.M.; et al. A Abd El-Wahed. Toxins 2021, 13, 206. [Google Scholar] [CrossRef]
  11. Mortari, M.R.; Cunha, A.O.S.; Ferreira, L.B.; dos Santos, W.F. Neurotoxins from invertebrates as anticonvulsants: From basic research to therapeutic application. Pharmacol. Ther. 2007, 114, 171–183. [Google Scholar] [CrossRef] [PubMed]
  12. El-Seedi, H.; El-Wahed, A.; Yosri, N.; Musharraf, S.G.; Chen, L.; Moustafa, M.; Zou, X.; Al-Mousawi, S.; Guo, Z.; Khatib, A. Antimicrobial properties of Apis mellifera’s bee venom. Toxins 2020, 12, 451. [Google Scholar] [CrossRef] [PubMed]
  13. Toledo, R.C.; Jared, C. Cutaneous granular glands and amphibian venoms. Comp. Biochem. Physiol.-Part A Physiol. 1995, 111, 1–29. [Google Scholar] [CrossRef]
  14. Van Bocxlaer, I.; Loader, S.P.; Roelants, K.; Biju, S.D.; Menegon, M.; Bossuyt, F. Gradual adaptation toward a range-expansion phenotype initiated the global radiation of toads. Science 2010, 327, 679–682. [Google Scholar] [CrossRef] [PubMed]
  15. Clarke, B.T. The natural history of amphibian skin secretions, their normal functioning and potential medical applications. Biol. Rev. Camb. Philos. Soc. 1997, 72, 365–379. [Google Scholar] [CrossRef]
  16. Soumoy, L.; Welles, M.; Najem, A.; Krayem, M.; Ghanem, G.; Hambye, S.; Saussez, S.; Blankert, B.; Journe, F. Toad Venom Antiproliferative Activities on Metastatic Melanoma: Bio-Guided Fractionation and Screening of the Compounds of Two Different Venoms. Biology 2020, 9, 218. [Google Scholar] [CrossRef]
  17. Tang, H.-J.; Ruan, L.-J.; Tian, H.-Y.; Liang, G.-P.; Ye, W.-C.; Hughes, E.; Esmann, M.; Fedosova, N.U.; Chung, T.-Y.; Tzen, J.T.C. Novel stereoselective bufadienolides reveal new insights into the requirements for Na+, K+-ATPase inhibition by cardiotonic steroids. Sci. Rep. 2016, 6, 29155. [Google Scholar] [CrossRef] [Green Version]
  18. Baldo, E.C.F.; Anjolette, F.A.P.; Arantes, E.C.; Baldo, M.A. Toad poison and drug discovery. Toxicon 2015, 16, 1–22. [Google Scholar]
  19. Perry, D. Proteins of parotoid gland secretions from toads of the genus Bufo. Contemp. Herpetol. 2000, 3, 1–7. [Google Scholar] [CrossRef]
  20. Jared, C.; Antoniazzi, M.M.; Jordão, A.E.C.; Silva, J.R.M.C.; Greven, H.; Rodrigues, M.T. Parotoid macroglands in toad (Rhinella jimi): Their structure and functioning in passive defence. Toxicon 2009, 54, 197–207. [Google Scholar] [CrossRef]
  21. Gopalakrishnakone, P.; Cruz, L.J.; Luo, S. Toxins and Drug Discovery; Gopalakrishnakone, P., Cruz, L.J., Luo, S., Eds.; Springer: Amsterdam, The Netherlands, 2015. [Google Scholar]
  22. de Sousa, L.Q.; da Conceição Machado, K.; de Carvalho Oliveira, S.F.; da Silva Araújo, L.; dos Santos Monção-Filho, E.; Amélia, A.; Vieira-Júnior, G.M.; Ferreira, P.M.P. Bufadienolides from amphibians: A promising source of anticancer prototypes for radical innovation, apoptosis triggering and Na+/K+-ATPase inhibition. Toxicon 2017, 127, 63–76. [Google Scholar] [CrossRef]
  23. Amarelle, L.; Lecuona, E. The Antiviral Effects of Na,K-ATPase Inhibition: A Minireview. Int. J. Mol. Sci. 2018, 19, 2154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Orellana, A.M.; Kinoshita, P.F.; Leite, J.A.; Kawamoto, E.M.; Scavone, C. Cardiotonic steroids as modulators of neuroinflammation. Front. Endocrinol. 2016, 7, 10. [Google Scholar] [CrossRef] [Green Version]
  25. Hayes, R.A.; Piggott, A.M.; Dalle, K.; Capon, R.J. Microbial biotransformation as a source of chemical diversity in cane toad steroid toxins. Bioorganic Med. Chem. Lett. 2009, 19, 1790–1792. [Google Scholar] [CrossRef] [PubMed]
  26. Üveges, B.; Fera, G.; Móricz, Á.M.; Krüzselyi, D.; Bókony, V.; Hettyey, A. Age- and environment-dependent changes in chemical defences of larval and post-metamorphic toads. BMC Evol. Biol. 2017, 17, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Bókony, V.; Üveges, B.; Móricz, Á.M.; Hettyey, A. Competition induces increased toxin production in toad larvae without allelopathic effects on heterospecific tadpoles. Funct. Ecol. 2018, 32, 667–675. [Google Scholar] [CrossRef] [Green Version]
  28. Bókony, V.; Móricz, Á.M.; Tóth, Z.; Gál, Z.; Kurali, A.; Mikó, Z.; Pásztor, K.; Szederkényi, M.; Tóth, Z.; Ujszegi, J.; et al. Variation in Chemical Defense Among Natural Populations of Common Toad, Bufo bufo, Tadpoles: The Role of Environmental Factors. J. Chem. Ecol. 2016, 42, 329–338. [Google Scholar] [CrossRef] [PubMed]
  29. Chiadao, C.; Osuch, M. V Biosynthesis of bufadienolides—3β-hydroxycholanates as precursors in Bufo marinus bufadienolides synthesis. Biochem. Pharmacol. 1969, 18, 1797–1802. [Google Scholar] [CrossRef]
  30. Siperstein, M.D.; Murray, A.W.; Titus, E. Biosynthesis of cardiotonic sterols from cholesterol in the toad, Bufo marinus. Arch. Biochem. Biophys. 1957, 67, 154–160. [Google Scholar] [CrossRef]
  31. Porto, A.M.; Baralle, F.E.; Gros, E.G. Biosynthesis of bufadienolides in toads: III—Experiments with [2-14C] mevalonic acid, [20-14C] 3β-hydroxy-5-pregnen-20-one and [20-14C] cholesterol. J. Steroid Biochem. 1972, 3, 11–17. [Google Scholar] [CrossRef]
  32. Delfino, G.; Nosi, D.; Giachi, F. Secretory granule-cytoplasm relationships in serous glands of anurans: Ultrastructural evidence and possible functional role. Toxicon 2001, 39, 1161–1171. [Google Scholar] [CrossRef]
  33. Duellman, W.; Trueb, L. Biology of amphibians; Johns Hopkins University Press: Baltimore, MD, USA, 1994; ISBN 9780801847806. [Google Scholar]
  34. Mailho-Fontana, P.L.; Antoniazzi, M.M.; Toledo, L.F.; Verdade, V.K.; Sciani, J.M.; Barbaro, K.C.; Pimenta, D.C.; Rodrigues, M.T.; Jared, C. Passive and active defense in toads: The parotoid macroglands in Rhinella marina and Rhaebo guttatus. J. Exp. Zool. Part A Ecol. Genet. Physiol. 2014, 321, 65–77. [Google Scholar] [CrossRef]
  35. Jin, L.; Quan, C.; Hou, X.; Fan, S. Potential pharmacological resources: Natural bioactive compounds from marine-derived fungi. Mar. Drugs 2016, 14, 76. [Google Scholar] [CrossRef] [Green Version]
  36. Antoniazzi, M.M.; Neves, P.R.; Mailho-Fontana, P.L.; Rodrigues, M.T.; Jared, C. Morphology of the parotoid macroglands in Phyllomedusa leaf frogs. J. Zool. 2013, 291, 42–50. [Google Scholar] [CrossRef]
  37. Regis-Alves, E.; Jared, S.G.S.; Maurício, B.; Mailho-Fontana, P.L.; Antoniazzi, M.M.; Fleury-Curado, M.C.; Brodie, E.D.; Jared, C. Structural cutaneous adaptations for defense in toad (Rhinella icterica) parotoid macroglands. Toxicon 2017, 137, 128–134. [Google Scholar] [CrossRef] [PubMed]
  38. Brodie, E.D.; Smatresk, N.J. The antipredator arsenal of fire salamanders: Spraying of secretions from highly pressurized dorsal skin glands. Herpetologica 1990, 46, 1–7. [Google Scholar]
  39. Mailho-Fontana, P.L.; Antoniazzi, M.M.; Rodrigues, I.; Sciani, J.M.; Pimenta, D.C.; Brodie, E.D.; Rodrigues, M.T.; Jared, C. Parotoid, radial, and tibial macroglands of the frog Odontophrynus cultripes: Differences and similarities with toads. Toxicon 2017, 129, 123–133. [Google Scholar] [CrossRef]
  40. Tempone, A.G.; Pimenta, D.C.; Lebrun, I.; Sartorelli, P.; Taniwaki, N.N.; de Andrade, H.F.; Antoniazzi, M.M.; Jared, C. Antileishmanial and antitrypanosomal activity of bufadienolides isolated from the toad Rhinella jimi parotoid macrogland secretion. Toxicon 2008, 52, 13–21. [Google Scholar] [CrossRef]
  41. Gustavo Tempone, A.; de Souza Carvalho Melhem, M.; Oliveira Prado, F.; Motoie, G.; Mitsuyoshi Hiramoto, R.; Maria Antoniazzi, M.; Fernando Baptista Haddad, C.; Jared, C. Amphibian Secretions for Drug Discovery Studies: A Search for New Antiparasitic and Antifungal Compounds. Lett. Drug Des. Discov. 2006, 4, 67–73. [Google Scholar] [CrossRef]
  42. Dmitrieva, R.I.; Doris, P.A. Cardiotonic steroids: Potential endogenous sodium pump ligands with diverse function. Exp. Biol. Med. 2002, 227, 561–569. [Google Scholar] [CrossRef]
  43. Ivanchina, N.V.; Kicha, A.A.; Stonik, V.A. Steroid glycosides from marine organisms. Steroids 2011, 76, 425–454. [Google Scholar] [CrossRef] [PubMed]
  44. Ali, M.S.; Saleem, M.; Yamdagni, R.; Ali, M.A. Steroid and antibacterial steroidal glycosides from marine green alga Codium iyengarii borgesen. Nat. Prod. Lett. 2002, 16, 407–413. [Google Scholar] [CrossRef] [PubMed]
  45. Saikia, S.; Kolita, B.; Dutta, P.P.; Dutta, D.J.; Neipihoi; Nath, S.; Bordoloi, M.; Quan, P.M.; Thuy, T.T.; Phuong, D.L.; et al. Marine steroids as potential anticancer drug candidates: In silico investigation in search of inhibitors of Bcl-2 and CDK-4/Cyclin D1. Steroids 2015, 102, 7–16. [Google Scholar] [CrossRef] [PubMed]
  46. Dos Santos Dias, A.C.; Couzinet-Mossion, A.; Ruiz, N.; Lakhdar, F.; Etahiri, S.; Bertrand, S.; Ory, L.; Roussakis, C.; Pouchus, Y.F.; Nazih, E.H.; et al. Steroids from marine-derived fungi: Evaluation of antiproliferative and antimicrobial activities of eburicol. Mar. Drugs 2019, 17, 372. [Google Scholar] [CrossRef] [Green Version]
  47. El-Seedi, H.R.; Khalifa, S.A.M.; Taher, E.A.; Farag, M.A.; Saeed, A.; Gamal, M.; Hegazy, M.E.F.; Youssef, D.; Musharraf, S.G.; Alajlani, M.M.; et al. Cardenolides: Insights from chemical structure and pharmacological utility. Pharmacol. Res. 2019, 141, 123–175. [Google Scholar] [CrossRef]
  48. Melero, C.P.; Medarde, M.; San Feliciano, A. A short review on cardiotonic steroids and their aminoguanidine analogues. Molecules 2000, 5, 51–81. [Google Scholar] [CrossRef] [Green Version]
  49. Boff, L.; Schreiber, A.; da Rocha Matos, A.; Del Sarto, J.; Brunotte, L.; Munkert, J.; Ottoni, F.M.; Ramos, G.S.; Kreis, W.; Braga, F.C.; et al. Semisynthetic cardenolides acting as antiviral inhibitors of influenza A virus replication by preventing polymerase complex formation. Molecules 2020, 25, 4853. [Google Scholar] [CrossRef]
  50. Offermanns, S.; Rosenthal, W. Encyclopedia of Molecular Pharmacology; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2008; ISBN 978-3-540-38916-3. [Google Scholar]
  51. Michalak, M.; Michalak, K.; Wicha, J. The synthesis of cardenolide and bufadienolide aglycones, and related steroids bearing a heterocyclic subunit. Nat. Prod. Rep. 2017, 34, 361–410. [Google Scholar] [CrossRef]
  52. Cornelius, F.; Kanai, R.; Toyoshima, C. A structural view on the functional importance of the sugar moiety and steroid hydroxyls of cardiotonic steroids in binding to Na,K-ATPase. J. Biol. Chem. 2013, 288, 6602–6616. [Google Scholar] [CrossRef] [Green Version]
  53. Morsy, N. Cardiac glycosides in medicinal plants, Aromatic and Medicinal Plants—Back to nature.; El-Shemy, H., Ed.; BoD—Books on Demand: London, UK, 2017; ISBN 9535129775/9789535129776. [Google Scholar]
  54. Dondoni, A.; Marra, A. Methods for Anomeric Carbon-Linked and Fused Sugar Amino Acid Synthesis:  The Gateway to Artificial Glycopeptides. Chem. Rev. 2000, 100, 4395–4422. [Google Scholar] [CrossRef]
  55. Křen, V. Glycoside vs. Aglycon: The Role of Glycosidic Residue in Biological Activity. In Glycoscience; Springer: Berlin, Heidelberg, 2008; pp. 2589–2644. [Google Scholar]
  56. Gupta, S.P. Quantitative structure-activity relationships of cardiotonic agents. Prog. Drug Res. 2000, 55, 235–282. [Google Scholar] [PubMed]
  57. Yang, Q.; Zhou, X.; Zhang, M.; Bi, L.; Miao, S.; Cao, W.; Xie, Y.; Sun, J.; Tang, H.; Li, Y.; et al. Angel of human health: Current research updates in toad medicine. Am. J. Transl. Res. 2015, 7, 1–14. [Google Scholar] [PubMed]
  58. Klupczynska, A.; Pawlak, M.; Kokot, Z.J.; Matysiak, J. Application of metabolomic tools for studying low molecular-weight fraction of animal venoms and poisons. Toxins 2018, 10, 306. [Google Scholar] [CrossRef] [Green Version]
  59. Kamano, Y.; Kotake, A.; Hashima, H.; Inoue, M.; Morita, H.; Takeya, K.; Itokawa, H.; Nandachi, N.; Segawa, T.; Yukita, A.; et al. Structure–cytotoxic activity relationship for the toad poison bufadienolides. Bioorg. Med. Chem. 1998, 6, 1103–1115. [Google Scholar] [CrossRef]
  60. Ye, M.; Qu, G.; Guo, H.; Guo, D. Specific 12β-hydroxylation of cinobufagin by filamentous fungi. Appl. Environ. Microbiol. 2004, 70, 3521–3527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Li, M.; Yu, X.; Guo, H.; Sun, L.; Wang, A.; Liu, Q.; Wang, X.; Li, J. Bufalin exerts antitumor effects by inducing cell cycle arrest and triggering apoptosis in pancreatic cancer cells. Tumor Biol. 2014, 35, 2461–2471. [Google Scholar] [CrossRef]
  62. Qian, L.; Su, H.; Wang, G.; Li, B.; Shen, G.; Gao, Q. Anti-tumor activity of bufalin by inhibiting c-MET mediated MEK/ERK and PI3K/AKT signaling pathways in gallbladder cancer. J. Cancer 2020, 11, 3114–3123. [Google Scholar] [CrossRef] [Green Version]
  63. Su, S.; Dou, H.; Wang, Z.; Zhang, Q. Bufalin inhibits ovarian carcinoma via targeting mTOR/HIF-α pathway. Basic Clin. Pharmacol. Toxicol. 2021, 128, 224–233. [Google Scholar] [CrossRef]
  64. Qi, J.; Zulfiker, A.H.M.; Li, C.; Good, D.; Wei, M.Q. The development of toad toxins as potential therapeutic agents. Toxins 2018, 10, 336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Cohen, T.; Williams, J.D.; Opperman, T.J.; Sanchez, R.; Lurain, N.S.; Tortorella, D. Convallatoxin-Induced Reduction of Methionine Import Effectively Inhibits Human Cytomegalovirus Infection and Replication. J. Virol. 2016, 90, 10715–10727. [Google Scholar] [CrossRef] [Green Version]
  66. Burkard, C.; Verheije, M.H.; Haagmans, B.L.; van Kuppeveld, F.J.; Rottier, P.J.M.; Bosch, B.-J.; de Haan, C.A.M. ATP1A1-Mediated Src Signaling Inhibits Coronavirus Entry into Host Cells. J. Virol. 2015, 89, 4434–4448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Laird, G.M.; Eisele, E.E.; Rabi, S.A.; Nikolaeva, D.; Siliciano, R.F. A novel cell-based high-throughput screen for inhibitors of HIV-1 gene expression and budding identifies the cardiac glycosides. J. Antimicrob. Chemother. 2014, 69, 988–994. [Google Scholar] [CrossRef] [PubMed]
  68. Dodson, A.W.; Taylor, T.J.; Knipe, D.M.; Coen, D.M. Inhibitors of the sodium potassium ATPase that impair herpes simplex virus replication identified via a chemical screening approach. Virology 2007, 366, 340–348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Zhang, Z.R.; Zhang, Y.N.; Li, X.D.; Zhang, H.Q.; Xiao, S.Q.; Deng, F.; Yuan, Z.M.; Ye, H.Q.; Zhang, B. A cell-based large-scale screening of natural compounds for inhibitors of SARS-CoV-2. Signal Transduct. Target. Ther. 2020, 5, 218. [Google Scholar] [CrossRef]
  70. Jin, Y.-H.; Jeon, S.; Lee, J.; Kim, S.; Jang, M.S.; Park, C.M.; Song, J.H.; Kim, H.R.; Kwon, S. Broad Spectrum Antiviral Properties of Cardiotonic Steroids Used as Potential Therapeutics for Emerging Coronavirus Infections. Pharmaceutics 2021, 13, 1839. [Google Scholar] [CrossRef] [PubMed]
  71. Cui, X.; Inagaki, Y.; Xu, H.; Wang, D.; Qi, F.; Kokudo, N.; Fang, D.; Tang, W. Anti-hepatitis B virus activities of cinobufacini and its active components bufalin and cinobufagin in HepG2.2.15 Cells. Biol. Pharm. Bull. 2010, 33, 1728–1732. [Google Scholar] [CrossRef] [Green Version]
  72. Chen, J.; Xu, L.; Sun, S.; Zhang, H.; Ma, T.; Su, W.; Jiang, C. Identification of cinobufagin and resibufogenin as inhibitors of enterovirus 71 infection. Chem. Res. Chinese Univ. 2014, 30, 953–958. [Google Scholar] [CrossRef]
  73. Wong, R.W.; Lingwood, C.A.; Ostrowski, M.A.; Cabral, T.; Cochrane, A. Cardiac glycoside/aglycones inhibit HIV-1 gene expression by a mechanism requiring MEK1/2-ERK1/2 signaling. Sci. Rep. 2018, 8, 850. [Google Scholar] [CrossRef] [Green Version]
  74. Horisberger, J.D. Recent insights into the structure and mechanism of the sodium pump. Physiology 2004, 19, 377–387. [Google Scholar] [CrossRef] [Green Version]
  75. Rahimtoola, S.H.; Tak, T. The use of digitalis in heart failure. Curr. Probl. Cardiol. 1996, 21, 781–853. [Google Scholar] [CrossRef]
  76. Weidemann, H. NA/K-ATPase, endogenous digitalis-like compounds and cancer development—A hypothesis. Front. Biosci. 2005, 10, 2165–2176. [Google Scholar] [CrossRef] [PubMed]
  77. Gonta-Grabiec, K.; Rossowski, W.; Szumiel, I. Properties of Na+/K+ ATPase and alkaline phosphatase alter during spontaneous and radiation-induced leukemogenesis in mice. Neoplasma 1986, 33, 141–155. [Google Scholar] [PubMed]
  78. Kaplan, J.G. Membrane cation transport and the control of proliferation of mammalian cells. Annu. Rev. Physiol. 1978, 40, 19–41. [Google Scholar] [CrossRef]
  79. Liu, S.H.; Lin, C.H.; Hung, S.K.; Chou, J.H.; Chi, C.W.; Fu, S.L. Fisetin inhibits lipopolysaccharide-induced macrophage activation and dendritic cell maturation. J. Agric. Food Chem. 2010, 58, 10831–10839. [Google Scholar] [CrossRef]
  80. Shen, S.S.; Hamamoto, S.T.; Bern, H.A.; Steinhardt, R.A. Alteration of Sodium Transport in Mouse Mammary Epithelium Associated with Neoplastic Transformation. Cancer Res. 1978, 38, 1356–1361. [Google Scholar] [PubMed]
  81. Huang, L.; Li, H.; Xie, Z. Ouabain-induced hypertrophy in cultured cardiac myocytes is accompanied by changes in expression of several late response genes. J. Mol. Cell. Cardiol. 1997, 29, 429–437. [Google Scholar] [CrossRef]
  82. Davies, R.J.; Sandle, G.I.; Thompson, S.M. Inhibition of the Na+,K(+)-ATPase pump during induction of experimental colon cancer. Cancer Biochem. Biophys. 1991, 12, 81–94. [Google Scholar]
  83. Kometiani, P.; Li, J.; Gnudi, L.; Kahn, B.B.; Askari, A.; Xie, Z. Multiple signal transduction pathways link Na+/K+-ATPase to growth- related genes in cardiac myocytes: The roles of Ras and mitogen-activated protein kinases. J. Biol. Chem. 1998, 273, 15249–15256. [Google Scholar] [CrossRef] [Green Version]
  84. Xie, Z.; Kometiani, P.; Liu, J.; Li, J.; Shapiro, J.I.; Askari, A. Intracellular reactive oxygen species mediate the linkage of Na+/K+-ATPase to hypertrophy and its marker genes in cardiac myocytes. J. Biol. Chem. 1999, 274, 19323–19328. [Google Scholar] [CrossRef] [Green Version]
  85. Haas, M.; Wang, H.; Tian, J.; Xie, Z. Src-mediated inter-receptor cross-talk between the Na+/K+-ATPase and the epidermal growth factor receptor relays the signal from ouabain to mitogen-activated protein kinases. J. Biol. Chem. 2002, 277, 18694–18702. [Google Scholar] [CrossRef] [Green Version]
  86. Cerella, C.; Dicato, M.; Diederich, M. Assembling the puzzle of anti-cancer mechanisms triggered by cardiac glycosides. Mitochondrion 2013, 13, 225–234. [Google Scholar] [CrossRef]
  87. Menger, L.; Vacchelli, E.; Kepp, O.; Eggermont, A.; Tartour, E.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Trial watch: Cardiac glycosides and cancer therapy. Oncoimmunology 2013, 2, e23082. [Google Scholar] [CrossRef] [Green Version]
  88. Haas, M.; Askari, A.; Xie, Z. Involvement of Src and epidermal growth factor receptor in the signal-transducing function of Na+/K+-ATPase. J. Biol. Chem. 2000, 275, 27832–27837. [Google Scholar] [CrossRef] [PubMed]
  89. Mekhail, T.; Kaur, H.; Ganapathi, R.; Budd, G.T.; Elson, P.; Bukowski, R.M. Phase 1 trial of AnvirzelTM in patients with refractory solid tumors. Invest. New Drugs 2006, 24, 423–427. [Google Scholar] [CrossRef]
  90. Feng, B.; Guo, Y.W.; Huang, C.G.; Li, L.; Chen, R.H.; Jiao, B.H. 2′-epi-2′-O-Acetylthevetin B extracted from seeds of Cerbera manghas L. induces cell cycle arrest and apoptosis in human hepatocellular carcinoma HepG2 cells. Chem. Biol. Interact. 2010, 183, 142–153. [Google Scholar] [CrossRef]
  91. Varbanov, H.P.; Kuttler, F.; Banfi, D.; Turcatti, G.; Dyson, P.J. Repositioning approved drugs for the treatment of problematic cancers using a screening approach. PLoS ONE 2017, 12, e0171052. [Google Scholar] [CrossRef] [Green Version]
  92. Liang, M.; Tian, J.; Liu, L.; Pierre, S.; Liu, J.; Shapiro, J.; Xie, Z.J. Identification of a pool of non-pumping Na/K-ATPase. J. Biol. Chem. 2007, 282, 10585–10593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Mijatovic, T.; Roland, I.; Van Quaquebeke, E.; Nilsson, B.; Mathieu, A.; Van Vynckt, F.; Darro, F.; Blanco, G.; Facchini, V.; Kiss, R. The α1 subunit of the sodium pump could represent a novel target to combat non-small cell lung cancers. J. Pathol. 2007, 212, 170–179. [Google Scholar] [CrossRef]
  94. Mathieu, V.; Pirker, C.; Martin de Lassalle, E.; Vernier, M.; Mijatovic, T.; DeNeve, N.; Gaussin, J.F.; Dehoux, M.; Lefranc, F.; Berger, W.; et al. The sodium pump α1 sub-unit: A disease progression-related target for metastatic melanoma treatment. J. Cell. Mol. Med. 2009, 13, 3960–3972. [Google Scholar] [CrossRef] [Green Version]
  95. Lefranc, F.; Kiss, R. The sodium pump α1 subunit as a potential target to combat apoptosis-resistant glioblastomas. Neoplasia 2008, 10, 198–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Kim, N.; Yim, H.Y.; He, N.; Lee, C.J.; Kim, J.H.; Choi, J.S.; Lee, H.S.; Kim, S.; Jeong, E.; Song, M.; et al. Cardiac glycosides display selective efficacy for STK11 mutant lung cancer. Sci. Rep. 2016, 6, 29721. [Google Scholar] [CrossRef] [Green Version]
  97. Yang, Y.; Cai, X.; Yang, J.; Sun, X.; Hu, C.; Yan, Z.; Xu, X.; Lu, W.; Wang, X.; Cao, P. Chemoprevention of dietary digitoflavone on colitis-associated colon tumorigenesis through inducing Nrf2 signaling pathway and inhibition of inflammation. Mol. Cancer 2014, 13, 48. [Google Scholar] [CrossRef] [Green Version]
  98. Wang, Y.; Qiu, Q.; Shen, J.J.; Li, D.D.; Jiang, X.J.; Si, S.Y.; Shao, R.G.; Wang, Z. Cardiac glycosides induce autophagy in human non-small cell lung cancer cells through regulation of dual signaling pathways. Int. J. Biochem. Cell Biol. 2012, 44, 1813–1824. [Google Scholar] [CrossRef]
  99. Juncker, T.; Schumacher, M.; Dicato, M.; Diederich, M. UNBS1450 from Calotropis procera as a regulator of signaling pathways involved in proliferation and cell death. Biochem. Pharmacol. 2009, 78, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Pongrakhananon, V.; Chunhacha, P.; Chanvorachote, P. Ouabain Suppresses the Migratory Behavior of Lung Cancer Cells. PLoS ONE 2013, 8, e68623. [Google Scholar] [CrossRef] [PubMed]
  101. Schneider, N.F.Z.; Geller, F.C.; Persich, L.; Marostica, L.L.; Pádua, R.M.; Kreis, W.; Braga, F.C.; Simões, C.M.O. Inhibition of cell proliferation, invasion and migration by the cardenolides digitoxigenin monodigitoxoside and convallatoxin in human lung cancer cell line. Nat. Prod. Res. 2016, 30, 1327–1331. [Google Scholar] [CrossRef]
  102. Yang, S.Y.; Kim, N.H.; Cho, Y.S.; Lee, H.; Kwon, H.J. Convallatoxin, a dual inducer of autophagy and apoptosis, inhibits angiogenesis in vitro and in vivo. PLoS ONE 2014, 9, e91094. [Google Scholar] [CrossRef]
  103. Watabe, M.; Masuda, Y.; Nakajo, S.; Yoshida, T.; Kuroiwa, Y.; Nakaya, K. The cooperative interaction of two different signaling pathways in response to bufalin induces apoptosis in human leukemia U937 cells. J. Biol. Chem. 1996, 271, 14067–14073. [Google Scholar] [CrossRef] [Green Version]
  104. Kurosawa, M.; Numazawa, S.; Tani, Y.; Yoshida, T. ERK signaling mediates the induction of inflammatory cytokines by bufalin in human monocytic cells. Am. J. Physiol.-Cell Physiol. 2000, 278, C500–C508. [Google Scholar] [CrossRef] [Green Version]
  105. Jiang, Y.; Zhang, Y.; Luan, J.; Duan, H.; Zhang, F.; Yagasaki, K.; Zhang, G. Effects of bufalin on the proliferation of human lung cancer cells and its molecular mechanisms of action. Cytotechnology 2010, 62, 573–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Watabe, M.; Ito, K.; Masuda, Y.; Nakajo, S.; Nakaya, K. Activation of AP-1 is required for bufalin-induced apoptosis in human leukemia U937 cells. Oncogene 1998, 16, 779–787. [Google Scholar] [CrossRef] [Green Version]
  107. Zhang, J.; Sha, J.; Zhou, Y.; Han, K.; Wang, Y.; Su, Y.; Yin, X.; Hu, H.; Yao, Y. Bufalin Inhibits Proliferation and Induces Apoptosis in Osteosarcoma Cells by Downregulating MicroRNA-221. Evid.-Based Complement. Altern. Med. 2016, 2016, 7319464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Yu, C.H.; Kan, S.F.; Pu, H.F.; Jea Chien, E.; Wang, P.S. Apoptotic signaling in bufalin- and cinobufagin-treated androgen-dependent and -independent human prostate cancer cells. Cancer Sci. 2008, 99, 2467–2476. [Google Scholar] [CrossRef]
  109. Yamada, K.; Hino, K.I.; Tomoyasu, S.; Honma, Y.; Tsuruoka, N. Enhancement by bufalin of retinoic acid-induced differentiation of acute promyelocytic leukemia cells in primary culture. Leuk. Res. 1998, 22, 589–595. [Google Scholar] [CrossRef]
  110. Qi, F.; Inagaki, Y.; Gao, B.; Cui, X.; Xu, H.; Kokudo, N.; Li, A.; Tang, W. Bufalin and cinobufagin induce apoptosis of human hepatocellular carcinoma cells via Fas- and mitochondria-mediated pathways. Cancer Sci. 2011, 102, 951–958. [Google Scholar] [CrossRef]
  111. Chen, X.Y.; Xu, R.C.; Chen, L. Apoptosis of gastric cancer cells induced by bufalin. Basic Med. 2000, 5, 50–54. [Google Scholar]
  112. Masuda, Y.; Kawazoe, N.; Nakajo, S.; Yoshida, T.; Kuroiwa, Y.; Nakaya, K. Bufalin induces apoptosis and influences the expression of apoptosis-related genes in human leukemia cells. Leuk. Res. 1995, 19, 549–556. [Google Scholar] [CrossRef]
  113. Watabe, M.; Kawazoe, N.; Masuda, Y.; Nakajo, S.; Nakaya, K. Bcl-2 protein inhibits bufalin-induced apoptosis through inhibition of mitogenactivated protein kinase activation in human leukemia U937 cells. Cancer Res. 1997, 57, 3097–3100. [Google Scholar]
  114. Han, K.Q.; Huang, G.; Gu, W.; Su, Y.H.; Huang, X.Q.; Ling, C.Q. Anti-tumor activities and apoptosis-regulated mechanisms of bufalin on the orthotopic transplantation tumor model of human hepatocellular carcinoma in nude mice. World J. Gastroenterol. 2007, 13, 3374–3379. [Google Scholar] [CrossRef]
  115. Zhang, J.; Zhou, X.; Zhou, Y.; Wang, Y.; Qian, B.; He, A.; Shen, Z.; Hu, H.; Yao, Y. Bufalin suppresses the migration and invasion of prostate cancer cells through HOTAIR, the sponge of miR-520b. Acta Pharmacol. Sin. 2019, 40, 1228–1236. [Google Scholar] [CrossRef] [PubMed]
  116. Li, Y.; Tian, X.; Liu, X.; Gong, P. Bufalin inhibits human breast cancer tumorigenesis by inducing cell death through the ROS-mediated RIP1/RIP3/PARP-1 pathways. Carcinogenesis 2018, 39, 700–707. [Google Scholar] [CrossRef] [Green Version]
  117. Zhen, S.; Hua, L.; Liu, Y.-H.; Wan, D.-Y.; Luo, W.-J. Bufalin attenuates the proliferation of breast cancer MCF-7 cells in vitro and in vivo by inhibiting the PI3K/Akt pathway. Int. J. Clin. Exp. Med. I 2016, 3, 10297–10303. [Google Scholar]
  118. Nakata, M.; Mori, S.; Kamoshida, Y.; Kawaguchi, S.; Fujita-Yamaguchi, Y.; Gao, B.; Tang, W. Toad skin extract cinobufatini inhibits migration of human breast carcinoma MDA-MB-231 cells into a model stromal tissue. Biosci. Trends 2015, 9, 266–269. [Google Scholar] [CrossRef] [PubMed]
  119. Dong, Y.; Yin, S.; Li, J.; Jiang, C.; Ye, M.; Hu, H. Bufadienolide compounds sensitize human breast cancer cells to TRAIL-induced apoptosis via inhibition of STAT3/Mcl-1 pathway. Apoptosis 2011, 16, 394–403. [Google Scholar] [CrossRef] [PubMed]
  120. Liu, M.; Feng, L.X.; Sun, P.; Liu, W.; Wu, W.Y.; Jiang, B.H.; Yang, M.; Hu, L.H.; Guo, D.A.; Liu, X. A novel bufalin derivative exhibited stronger apoptosis-inducing effect than bufalin in A549 lung cancer cells and lower acute toxicity in mice. PLoS ONE 2016, 11, e0159789. [Google Scholar] [CrossRef] [Green Version]
  121. Zhu, Z.; Sun, H.; Ma, G.; Wang, Z.; Li, E.; Liu, Y.; Liu, Y. Bufalin induces lung cancer cell apoptosis via the inhibition of PI3K/Akt pathway. Int. J. Mol. Sci. 2012, 13, 2025–2035. [Google Scholar] [CrossRef]
  122. Pan, Z.; Xie, Y.; Bai, J.; Lin, Q.; Cui, X.; Zhang, N. Bufalin suppresses colorectal cancer cell growth through promoting autophagy: In vivo and in vitro. RSC Adv. 2018, 8, 38910–38918. [Google Scholar] [CrossRef] [Green Version]
  123. Zhang, N.; Xie, Y.; Tai, Y.; Gao, Y.; Guo, W.; Yu, W.; Li, J.; Feng, X.; Hao, J.; Gao, Y.; et al. Bufalin Inhibits hTERT Expression and Colorectal Cancer Cell Growth by Targeting CPSF4. Cell. Physiol. Biochem. 2016, 40, 1559–1569. [Google Scholar] [CrossRef]
  124. Yuan, Z.T.; Shi, X.J.; Yuan, Y.X.; Qiu, Y.Y.; Zou, Y.; Liu, C.; Yu, H.; He, X.; Xu, K.; Yin, P.H. Bufalin reverses ABCB1-mediated drug resistance in colorectal cancer. Oncotarget 2017, 8, 48012–48026. [Google Scholar] [CrossRef] [Green Version]
  125. Qi, H.Y.; Qu, X.J.; Liu, J.; Hou, K.Z.; Fan, Y.B.; Che, X.F.; Liu, Y.P. Bufalin induces protective autophagy by Cbl-b regulating mTOR and ERK signaling pathways in gastric cancer cells. Cell Biol. Int. 2019, 43, 33–43. [Google Scholar] [CrossRef] [Green Version]
  126. Zhao, H.; Zhao, D.; Tan, G.; Liu, Y.; Zhuang, L.; Liu, T. Bufalin promotes apoptosis of gastric cancer by down-regulation of miR-298 targeting bax. Int. J. Clin. Exp. Med. 2015, 8, 3420. [Google Scholar] [PubMed]
  127. Miao, Q.; Bi, L.L.; Li, X.; Miao, S.; Zhang, J.; Zhang, S.; Yang, Q.; Xie, Y.H.; Zhang, J.; Wang, S.W. Anticancer effects of bufalin on human hepatocellular carcinoma HepG2 Cells: Roles of apoptosis and autophagy. Int. J. Mol. Sci. 2013, 14, 1370–1382. [Google Scholar] [CrossRef] [PubMed]
  128. Schoner, W. Endogenous cardiac glycosides, a new class of steroid hormones. Eur. J. Biochem. 2002, 269, 2440–2448. [Google Scholar] [CrossRef]
  129. Schoner, W.; Scheiner-Bobis, G. Endogenous cardiac glycosides: Hormones using the sodium pump as signal transducer. Semin. Nephrol. 2005, 25, 343–351. [Google Scholar] [CrossRef] [PubMed]
  130. Schoner, W.; Scheiner-Bobis, G. Endogenous and exogenous cardiac glycosides: Their roles in hypertension, salt metabolism, and cell growth. Am. J. Physiol.-Cell Physiol. 2007, 293, C509–C536. [Google Scholar] [CrossRef] [PubMed]
  131. Fedorova, O.V.; Talan, M.I.; Agalakova, N.I.; Lakatta, E.G.; Bagrov, A.Y. Endogenous ligand of α1 sodium pump, marinobufagenin, is a novel mediator of sodium chloride-dependent hypertension. Circulation 2002, 105, 1122–1127. [Google Scholar] [CrossRef] [PubMed]
  132. Uddin, M.N.; Allen, S.R.; Jones, R.O.; Zawieja, D.C.; Kuehl, T.J. Pathogenesis of pre-eclampsia: Marinobufagenin and angiogenic imbalance as biomarkers of the syndrome. Transl. Res. 2012, 160, 99–113. [Google Scholar] [CrossRef] [PubMed]
  133. Puschett, J.B.; Agunanne, E.; Uddin, M.N. Marinobufagenin, resibufogenin and preeclampsia. Biochim. Biophys. Acta-Mol. Basis Dis. 2010, 1802, 1246–1253. [Google Scholar] [CrossRef] [Green Version]
  134. Bagrov, A.Y.; Fedorova, O.V.; Dmitrieva, R.I.; Howald, W.N.; Hunter, A.P.; Kuznetsova, E.A.; Shpen, V.M. Characterization of a urinary bufodienolide Na+, K+-ATPase inhibitor in patients after acute myocardial infarction. Hypertension 1998, 31, 1097–1103. [Google Scholar] [CrossRef] [Green Version]
  135. Bagrov, A.Y.; Roukoyatkina, N.I.; Pinaev, A.G.; Dmitrieva, R.I.; Fedorova, O.V. Effects of two endogenous Na+, K+-ATPase inhibitors, marinobufagenin and ouabain, on isolated rat aorta. Eur. J. Pharmacol. 1995, 274, 151–158. [Google Scholar] [CrossRef]
  136. Fedorova, O.V.; Doris, P.A.; Bagrov, A.Y. Endogenous marinobufagenin-like factor in acute plasma volume expansion. Clin. Exp. Hypertens. 1998, 20, 581–591. [Google Scholar] [CrossRef] [PubMed]
  137. Bagrov, A.Y.; Fedorova, O.V.; Dmitriev, R.I.; French, A.W.; Anderson, D.E. Plasma marinobufagenin-like and ouabain-like immunoreactivity during saline volume expansion in anesthetized dogs. Cardiovasc. Res. 1996, 31, 296–305. [Google Scholar] [CrossRef] [Green Version]
  138. Hauck, C.; Frishman, W.H. Systemic hypertension: The roles of salt, vascular Na+/K+ ATPase and the endogenous glycosides, ouabain and marinobufagenin. Cardiol. Rev. 2012, 20, 130–138. [Google Scholar] [CrossRef] [PubMed]
  139. Gonick, H.C.; Ding, Y.; Vaziri, N.D.; Bagrov, A.Y.; Fedorova, O.V. Simultaneous Measurement of Marinobufagenin, Ouabain, and Hypertension-associated Protein In Various Disease States. Clin. Exp. Hypertens. 1998, 20, 617–627. [Google Scholar] [CrossRef] [PubMed]
  140. Lopatin, D.A.; Ailamazian, E.K.; Dmitrieva, R.I.; Shpen, V.M.; Fedorova, O.V.; Doris, P.A.; Bagrov, A.Y. Circulating bufodienolide and cardenolide sodium pump inhibitors in preeclampsia. J. Hypertens. 1999, 17, 1179–1187. [Google Scholar] [CrossRef] [PubMed]
  141. Lan, Y.L.; Wang, X.; Lou, J.C.; Xing, J.S.; Zou, S.; Yu, Z.L.; Ma, X.C.; Wang, H.; Zhang, B. Marinobufagenin inhibits glioma growth through sodium pump α1 subunit and ERK signaling-mediated mitochondrial apoptotic pathway. Cancer Med. 2018, 7, 2034–2047. [Google Scholar] [CrossRef]
  142. Chu, Q.; Xu, H.; Gao, M.; Guan, X.; Liu, H.; Deng, S.; Huo, X.; Liu, K.; Tian, Y.; Ma, X. Liver-targeting resibufogenin-loaded poly(Lactic-co-glycolic acid)-D-α-tocopheryl polyethylene glycol 1000 succinate nanoparticles for liver cancer therapy. Int. J. Nanomed. 2016, 11, 449–463. [Google Scholar]
  143. Lu, Z.; Xu, A.; Yuan, X.; Chen, K.; Wang, L.; Guo, T. Anticancer effect of resibufogenin on gastric carcinoma cells through the phosphoinositide 3-kinase/protein kinase B/glycogen synthase kinase 3β signaling pathway. Oncol. Lett. 2018, 16, 3297–3302. [Google Scholar] [CrossRef] [Green Version]
  144. Han, Q.; Ma, Y.; Wang, H.; Dai, Y.; Chen, C.; Liu, Y.; Jing, L.; Sun, X. Resibufogenin suppresses colorectal cancer growth and metastasis through RIP3-mediated necroptosis. J. Transl. Med. 2018, 16, 201. [Google Scholar] [CrossRef] [Green Version]
  145. Liu, L.; Liu, Y.; Liu, X.; Zhang, N.; Mao, G.; Zeng, Q.; Yin, M.; Song, D.; Deng, H. Resibufogenin suppresses transforming growth factor-β-activated kinase 1-mediated nuclear factor-κB activity through protein kinase C-dependent inhibition of glycogen synthase kinase 3. Cancer Sci. 2018, 109, 3611–3622. [Google Scholar] [CrossRef]
  146. Zhang, D.M.; Liu, J.S.; Deng, L.J.; Chen, M.F.; Yiu, A.; Cao, H.H.; Tian, H.Y.; Fung, K.P.; Kurihara, H.; Pan, J.X.; et al. Arenobufagin, a natural bufadienolide from toad venom, induces apoptosis and autophagy in human hepatocellular carcinoma cells through inhibition of PI3K/Akt/mTOR pathway. Carcinogenesis 2013, 34, 1331–1342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Wu, J.-H.; Cao, Y.-T.; Pan, H.-Y.; Wang, L.-H. Identification of Antitumor Constituents in Toad Venom by Spectrum-Effect Relationship Analysis and Investigation on Its Pharmacologic Mechanism. Molecules 2020, 25, 4269. [Google Scholar] [CrossRef] [PubMed]
  148. Zhao, J.; Zhang, Q.; Zou, G.; Gao, G.; Yue, Q. Arenobufagin, isolated from toad venom, inhibited epithelial-to-mesenchymal transition and suppressed migration and invasion of lung cancer cells via targeting IKKβ/NFκB signal cascade. J. Ethnopharmacol. 2020, 250, 112492. [Google Scholar] [CrossRef] [PubMed]
  149. Zhang, D.M.; Liu, J.S.; Tang, M.K.; Yiu, A.; Cao, H.H.; Jiang, L.; Yuet-Wa Chan, J.; Tian, H.Y.; Fung, K.P.; Ye, W.C. Bufotalin from Venenum Bufonis inhibits growth of multidrug resistant HepG2 cells through G2/M cell cycle arrest and apoptosis. Eur. J. Pharmacol. 2012, 692, 19–28. [Google Scholar] [CrossRef]
  150. Wei, X.; He, J.; Gao, B.; Han, L.; Mao, Y.; Zhao, H.; Si, N.; Wang, H.; Yang, J.; Bian, B. Hellebrigenin anti-pancreatic cancer effects based on apoptosis and autophage. PeerJ 2020, 2020, e9011. [Google Scholar] [CrossRef]
  151. Deng, L.-J.; Hu, L.-P.; Peng, Q.-L.; Yang, X.-L.; Bai, L.-L.; Yiu, A.; Li, Y.; Tian, H.-Y.; Ye, W.-C.; Zhang, D.-M. Hellebrigenin induces cell cycle arrest and apoptosis in human hepatocellular carcinoma HepG2 cells through inhibition of Akt. Chem. Biol. Interact. 2014, 219, 184–194. [Google Scholar] [CrossRef]
  152. Yu, Z.; Guo, W.; Ma, X.; Zhang, B.; Dong, P.; Huang, L.; Wang, X.; Wang, C.; Huo, X.; Yu, W. Gamabufotalin, a bufadienolide compound from toad venom, suppresses COX-2 expression through targeting IKKβ/NF-κB signaling pathway in lung cancer cells. Mol. Cancer 2014, 13, 203. [Google Scholar] [CrossRef]
  153. Rong, X.; Ni, W.; Liu, Y.; Wen, J.; Qian, C.; Sun, L.; Wang, J. Bufalin, a bioactive component of the Chinese medicine Chansu, inhibits inflammation and invasion of human rheumatoid arthritis fibroblast-like synoviocytes. Inflammation 2014, 37, 1050–1058. [Google Scholar] [CrossRef]
  154. Yang, Q.; Huang, W.; Jozwik, C.; Lin, Y.; Glasman, M.; Caohuy, H.; Srivastava, M.; Esposito, D.; Gillette, W.; Hartley, J.; et al. Cardiac glycosides inhibit TNF-α/NF-κB signaling by blocking recruitment of TNF receptor-associated death domain to the TNF receptor. Proc. Natl. Acad. Sci. USA. 2005, 102, 9631–9636. [Google Scholar] [CrossRef] [Green Version]
  155. Carvalho, D.C.M.; Cavalcante-Silva, L.H.A.; De A Lima, É.; Galvão, J.G.F.M.; De A Alves, A.K.; Feijó, P.R.O.; Quintas, L.E.M.; Rodrigues-Mascarenhas, S. Marinobufagenin inhibits neutrophil migration and proinflammatory cytokines. J. Immunol. Res. 2019, 2019, 1094520. [Google Scholar] [CrossRef] [Green Version]
  156. Qi, F.; Li, A.; Inagaki, Y.; Kokudo, N.; Tamura, S.; Nakata, M.; Tang, W. Antitumor activity of extracts and compounds from the skin of the toad Bufo bufo gargarizans Cantor. Int. Immunopharmacol. 2011, 11, 342–349. [Google Scholar] [CrossRef] [PubMed]
  157. Wen, L.; Huang, Y.; Xie, X.; Huang, W.; Yin, J.; Lin, W.; Jia, Q.; Zeng, W. Anti-inflammatory and antinociceptive activities of bufalin in rodents. Mediators Inflamm. 2014, 2014, 171839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Ye, J.; Chen, S.; Maniatis, T. Cardiac glycosides are potent inhibitors of interferon-β gene expression. Nat. Chem. Biol. 2010, 7, 25–33. [Google Scholar] [CrossRef] [Green Version]
  159. Xie, X.B.; Yin, J.Q.; Wen, L.L.; Gao, Z.H.; Zou, C.Y.; Wang, J.; Huang, G.; Tang, Q.L.; Colombo, C.; He, W.L.; et al. Critical Role of Heat Shock Protein 27 in Bufalin-Induced Apoptosis in Human Osteosarcomas: A Proteomic-Based Research. PLoS ONE 2012, 7, e47375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Rahman, A.; Fazal, F. Blocking NF-kB: An inflammatory issue. Proc. Am. Thorac. Soc. 2011, 8, 497–503. [Google Scholar] [CrossRef] [PubMed]
  161. Ihenetu, K.; Espinosa, R.; De Leon, R.; Planas, G.; Perez-Pinero, A.; Waldbeser, L. Digoxin and digoxin-like immunoreactive factors (DLIF) modulate the release of pro-inflammatory cytokines. Inflamm. Res. 2008, 57, 519–523. [Google Scholar] [CrossRef] [PubMed]
  162. Jacob, P.L.; Leite, J.A.; Alves, A.K.A.; Rodrigues, Y.K.S.; Amorim, F.M.; Néris, P.L.N.; Oliveira, M.R.; Rodrigues-Mascarenhas, S. Immunomodulatory activity of ouabain in Leishmania leishmania amazonensis-infected Swiss mice. Parasitol. Res. 2013, 112, 1313–1321. [Google Scholar] [CrossRef]
  163. Huang, Y.; Yang, G.; Fei, J.; Wu, Y.; Yan, J. Bufotalin ameliorates experimental Sjögren’s syndrome development by inhibiting Th17 generation. Naunyn. Schmiedebergs. Arch. Pharmacol. 2020, 393, 1977–1985. [Google Scholar] [CrossRef]
  164. Zheng, Y.; Deng, L.; Cao, H.; Xu, N.; Zhang, D.; Tian, H.; Li, B.; Lu, Z.; Ye, W.; Yu, L. Screening of bufadienolides from toad venom identifies gammabufotalin as a potential anti-inflammatory agent. Planta Med. 2022, 88, 43–52. [Google Scholar] [CrossRef]
  165. Takai, N.; Kira, N.; Ishii, T.; Yoshida, T.; Nishida, M.; Nishida, Y.; Nasu, K.; Narahara, H. Bufalin, a traditional oriental medicine, induces apoptosis in human cancer cells. Asian Pacific J. Cancer Prev. 2012, 13, 399–402. [Google Scholar] [CrossRef] [Green Version]
  166. Zhang, Z.-J.; Yang, Y.-K.; Wu, W.-Z. Bufalin attenuates the stage and metastatic potential of hepatocellular carcinoma in nude mice. J. Transl. Med. 2014, 12, 57. [Google Scholar] [CrossRef] [Green Version]
  167. Wu, S.-H.; Bau, D.-T.; Hsiao, Y.-T.; Lu, K.-W.; Hsia, T.-C.; Lien, J.-C.; Ko, Y.-C.; Hsu, W.-H.; Yang, S.-T.; Huang, Y.-P.; et al. Bufalin induces apoptosis in vitro and has Antitumor activity against human lung cancer xenografts in vivo. Environ. Toxicol. 2017, 32, 1305–1317. [Google Scholar] [CrossRef]
  168. Yin, P.-H.; Liu, X.; Qiu, Y.-Y.; Cai, J.; Qin, J.; Zhu, H.-R.; Li, Q. Anti-tumor activity and apoptosis-regulation mechanisms of bufalin in various cancers: New hope for cancer patients. Asian Pacific J. cancer Prev. 2012, 13, 5339–5343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Karin, M. NF-κB as a Critical Link between Inflammation and Cancer. Cold Spring Harb. Perspect. Biol. 2009, 1, a000141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. (A) The basic steroid aglycone unit of cardiotonic steroids and their types, cardenolides and bufadienolides; (B) numbered chemical structure of the bufadienolide aglycone or steroidal nucleus, i.e., the main type of cardiotonic steroids (n denotes the number of the sugar monomer in the cardiac glycoside structures (n = 1–3)).
Figure 1. (A) The basic steroid aglycone unit of cardiotonic steroids and their types, cardenolides and bufadienolides; (B) numbered chemical structure of the bufadienolide aglycone or steroidal nucleus, i.e., the main type of cardiotonic steroids (n denotes the number of the sugar monomer in the cardiac glycoside structures (n = 1–3)).
Molecules 27 06586 g001
Figure 2. Proposed mechanism of the biosynthesis of bufadienolides by toads (Bufo marinus) [29].
Figure 2. Proposed mechanism of the biosynthesis of bufadienolides by toads (Bufo marinus) [29].
Molecules 27 06586 g002
Figure 3. Proposed mechanism of the biosynthesis of marinobufagenin by toads (Bufo marinus) [30].
Figure 3. Proposed mechanism of the biosynthesis of marinobufagenin by toads (Bufo marinus) [30].
Molecules 27 06586 g003
Figure 4. Chemical structures of most common compounds isolated from venom of toads.
Figure 4. Chemical structures of most common compounds isolated from venom of toads.
Molecules 27 06586 g004aMolecules 27 06586 g004b
Table 1. Antiviral activities of cardiotonic steroids (CTS) from different toad species.
Table 1. Antiviral activities of cardiotonic steroids (CTS) from different toad species.
Toad SpeciesCTSTargetRef.
Bufo gargarizans CantorBufalinAnti-hepatitis B above 10-2 µM
Anti-HIV, IC50 = 5 nm
Anti-MERS-CoV in vero cells
(IC50 = 0.018 µM)
after 24 h
Anti-MERS-CoV
Calu-3 human lung cells (IC50 = 0.544 µM)
(in vitro)
[70,71,73]
Bufo gargarizansCinobufaginAnti-hepatitis B above 10-1 µM
Anti- enterovirus 71 (EV-71)
IC50 = 10.94 nmol/L
Anti-HIV, IC50 = 24 nm
Anti-MERS-CoV in vero cells
IC50 = 0.017 µM
after 24 h
Anti-MERS-CoV
Calu-3 human lung cells
IC50 = 0.616 µM
(in vitro)
[70,71,72,73]
Bufo gargarizansResibufogeninAnti-enterovirus 71 (EV71)
IC50 = 218 nmol/L
Anti-MERS-CoV in vero cells
(IC50 = 1.612 µM)
after 24 h
Anti-MERS-CoV
Calu-3 human lung cells
IC50 = 15.970 µM
(in vitro)
[70,72]
Bufo gargarizansTelocinobufaginAnti-MERS-CoV in vero cells
IC50 = 0.027 µM
after 24 h
Anti-MERS-CoV
Calu-3 human lung cells
IC50 = 0.465 µM
(in vitro)
[70]
Bufo gargarizans CantorBufotalinAnti-MERS-CoV in vero cells
IC50 = 0.063 µM
Anti-MERS-CoV
Calu-3 human lung cells
IC50 = 1.630 µM
(in vitro)
[70]
Bufo gargarizans CantorCinobufotalinAnti-MERS-CoV in vero cells
IC50 = 0.23 µM
after 24 h
Anti-MERS-CoV
Calu-3 human lung cells
IC50 = 3.958 µM
(in vitro)
[70]
Table 2. Anticancer activities of cardiotonic steroids (CTS) from different toad species.
Table 2. Anticancer activities of cardiotonic steroids (CTS) from different toad species.
Toad SpeciesDetected CTSTarget Cell and Mechanism of ActionRef.
Bufo gargarizans Cantor/
Bufo melanostictus Suhneider
(Toad venom)
ArenobufaginHepG2 cells
(IC50 = 20.24 ± 3.84 nM)
HepG2/ADM cells
(IC50 = 7.46 ± 2.89 nM)
HL-60 cells
(IC50 = 27.70 ± 8.77 nM)
after 72 h
Induces apoptosis and autophagy, inhibition of the PI3K/Akt/mTOR pathway
(in vitro, in vivo)
[146]
Bufo gargarizansArenobufaginNon-small cell lung cancer cell (A549)
IC50 = 12.530 ng/mL
after 72 h
Induces apoptosis in A549 cells with the enhanced expression of cleaved PARP (poly ADP-ribose polymerase
(in vitro)
[147]
Bufo melanostictus SchneiderArenobufaginLung cancer (A549)
At (0.5, 1 and 2 nM) inhibited the mobility of A549 cells (59.9%, 41.1%, and 24.7%, respectively)
At (0.5, 1, and 2 nM) inhibited the mobility of H1299 cells (72.3%, 47.4%, and 22.4%, respectively)
after 48 h
Target IKKβ to inactive NFκB signaling cascade and change protein expression related to EMT
(in vitro and in vivo)
[148]
Bufo gargarizans CantorBufalinNon-small cell lung cancer NSCLC A549 cells
At 2.5–10 µM, bufalin- induced apoptosis and cell cycle arrest in G1 phase
[105,149]
HepG2 cell (IC50 = 0.61 ± 0.06 µM)
R-HepG2 cells (IC50 = 0.24 ± 0.02 µM)
Induces cell cycle arrest at G2/M phase
after 48 h
(in vitro)
Bufo gargarizans Cantor and Bufo melanostictus SchneiderBufotalinHepG2 cell (IC50 = 0.43 ± 0.07 µM)
R-HepG2 cells(IC50 = 0.13 ± 0.01 µM)
Induces cell cycle arrest at G2/M phase
after 48 h
(in vitro)
[149]
Bufo gargarizans Cantor and Bufo melanostictus SchneiderHellebrigeninHepG2 cells
(IC50 = 0.40 ± 0.05 µmol/L)
After 24 h
(IC50 = 0.13 ± 0.01µmol/L)
After 48 h
(IC50 = 0.10 ± 0.01 µmol/L)
After 72 h
Induces cell cycle arrest at G2/M phase
(in vitro)
[150,151]
Bufo gargarizans CantorGamabufotalin (CS-6)NSCLC (IC50 = 50 nM)
Inhibit NSCLC cells growth and enhance apoptosis induction
(in vitro)
[152]
Bufo gargarizansCinobufatolinH157 cancer cells
IC50 = 131.12 ng/mL
A549 cancer cells
IC50 = 23.08 ng/mL
after 72 h
(in vitro)
[147,149]
HepG2 cell (IC50 = 1.58 ± 0.21 µM)
R-HepG2 cells(IC50 = 0.74 ± 0.07 µM)
Induces cell cycle arrest at G2/M phase
after 48 h
(in vitro)
Bufo gargarizansTelocinobufaginH157 cancer cells
IC50 = 23.60 ng/mL
A549 cancer cells
IC50 = 27.882 ng/mL
after 72 h
(in vitro)
[147,149]
HepG2 cell (IC50 = 1.28 ± 0.19 µM)
R-HepG2 cells(IC50 = 0.49 ± 0.05 µM)
after 48 h
Induces cell cycle arrest at G2/M phase
(in vitro)
Bufo gargarizansResibufogeninGastric carcinoma cells (MGC-803)
(4 and 8 µM)
for 24 h and 48 h
Increased Bax/Bcl-2 expression, and suppressed cyclin D1, cyclin E, PI3K, phosphorylated AKT, phosphorylated GSK3β, and β-catenin protein expression in MGC-803 cells.
(in vitro)
[143]
Table 3. Anti-inflammatory activities of cardiotonic steroids (CTS) from different toad species.
Table 3. Anti-inflammatory activities of cardiotonic steroids (CTS) from different toad species.
Toad SpeciesDetected CTSActivityRef.
Rhinella schneideriMarinobufageninAnti-inflammatory
(10, 100, 1000, and 10,000 nM),
decreased IL-1β, IL-6, and TNF-α levels
(in vitro, in vivo)
[155]
Bufo gargarizans CantorBufotalinAnti-inflammatory against chronic inflammatory autoimmune disease
100 μg/kg in vivo and 200 nM in vitro
inhibiting proinflammatory Th17 population and secretion of inflammatory cytokines
[163]
Bufo gargarizans CantorBufalinAnti-inflammatory against carrageenan-induced paw edema model
(0.3 and 0.6 mg/kg, i.p.)
Downregulation of expression levels of nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), 1β (IL-1β), (IL-6), (TNF-α), and inhibited the activation of NF-κB signaling
(in vivo)
[157]
Bufo gargarizansGammabufotalin Anti-inflammatory
(1, 4, and 12 (50 μM); 2, 13, and 14 (10 μM); 3 and 6 (5 μM); 5 and 8 (1 μM); 7, 9, and 11 (0.5 μM); 10 (4 μM))
Inhibits LPS-induced inflammation
by suppressing myeloid differentiation primary response 88/nuclear factor-kappa B and STAT3 signal pathways.
(in vivo)
[164]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

El-Seedi, H.R.; Yosri, N.; El-Aarag, B.; Mahmoud, S.H.; Zayed, A.; Du, M.; Saeed, A.; Musharraf, S.G.; El-Garawani, I.M.; Habib, M.R.; et al. Chemistry and the Potential Antiviral, Anticancer, and Anti-Inflammatory Activities of Cardiotonic Steroids Derived from Toads. Molecules 2022, 27, 6586. https://doi.org/10.3390/molecules27196586

AMA Style

El-Seedi HR, Yosri N, El-Aarag B, Mahmoud SH, Zayed A, Du M, Saeed A, Musharraf SG, El-Garawani IM, Habib MR, et al. Chemistry and the Potential Antiviral, Anticancer, and Anti-Inflammatory Activities of Cardiotonic Steroids Derived from Toads. Molecules. 2022; 27(19):6586. https://doi.org/10.3390/molecules27196586

Chicago/Turabian Style

El-Seedi, Hesham R., Nermeen Yosri, Bishoy El-Aarag, Shaymaa H. Mahmoud, Ahmed Zayed, Ming Du, Aamer Saeed, Syed G. Musharraf, Islam M. El-Garawani, Mohamed R. Habib, and et al. 2022. "Chemistry and the Potential Antiviral, Anticancer, and Anti-Inflammatory Activities of Cardiotonic Steroids Derived from Toads" Molecules 27, no. 19: 6586. https://doi.org/10.3390/molecules27196586

Article Metrics

Back to TopTop