Next Article in Journal
Dairy Propionibacteria: Probiotic Properties and Their Molecular Bases
Previous Article in Journal
Pre-Amplification of Cell-Free DNA: Balancing Amplification Errors with Enhanced Sensitivity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiac Glycosides: From Natural Defense Molecules to Emerging Therapeutic Agents

by
Arturo Ponce
*,
Catalina Flores-Maldonado
and
Ruben G. Contreras
Department of Physiology, Biophysics and Neurosciences, CINVESTAV-Instituto Politecnico Nacional, Mexico City 07360, Mexico
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(6), 885; https://doi.org/10.3390/biom15060885
Submission received: 7 May 2025 / Revised: 14 June 2025 / Accepted: 15 June 2025 / Published: 17 June 2025
(This article belongs to the Section Natural and Bio-derived Molecules)

Abstract

Cardiac glycosides (CGs), a class of plant- and animal-derived compounds historically used to treat heart failure, have garnered renewed interest for their diverse pharmacological properties beyond Na+/K+-ATPase (NKA) inhibition. Recent studies reveal that CGs modulate key signaling pathways—such as NF-κB, PI3K/Akt, JAK/STAT, and MAPK—affecting processes central to cancer, viral infections, immune regulation, and neurodegeneration. In cancer, CGs induce multiple forms of regulated cell death, including apoptosis, ferroptosis, pyroptosis, and immunogenic cell death, while also inhibiting angiogenesis, epithelial–mesenchymal transition, and cell cycle progression. They demonstrate broad-spectrum antiviral activity by disrupting viral entry, replication, and mRNA processing in viruses such as HSV, HIV, influenza, and SARS-CoV-2. Immunologically, CGs regulate Th17 differentiation via RORγ signaling, although both inhibitory and agonistic effects have been reported. In the nervous system, CGs modulate neuroinflammation, support synaptic plasticity, and improve cognitive function in models of Alzheimer’s disease, epilepsy, and multiple sclerosis. Despite their therapeutic potential, clinical translation is hindered by narrow therapeutic indices and systemic toxicity. Advances in drug design and nanocarrier-based delivery are critical to unlocking CGs’ full potential as multi-target agents for complex diseases. This review synthesizes the current knowledge on the emerging roles of CGs and highlights strategies for their safe and effective repurposing.

1. Introduction

Cardiac glycosides (CGs) are a structurally diverse class of naturally derived compounds. Found as secondary metabolites in various flowering plants, they are also produced by certain animals as a means of predator deterrence. The term cardiac glycosides stems from their characteristic chemical framework and their historically established use as cardiotonic drugs [1,2].
Throughout history, humans have recognized both the medicinal and toxic properties of CG-producing organisms. Some species were avoided due to their harmful effects, while others were deliberately used as poisons in hunting and warfare. CGs also found applications in traditional medicine. For example, the ancient Egyptians extracted CGs from Nerium oleander to treat heart conditions [3], while Digitalis purpurea (foxglove) was historically administered to manage edema. Traditional Chinese medicine likewise incorporates CGs; Chansu—derived from toad skin and venom—has been used for centuries to treat inflammatory disorders and certain cancers [4,5].
The cardiotonic properties of CGs were first scientifically documented in the late 18th century by the English physician William Withering, who demonstrated the efficacy of digitalis in treating dropsy by improving and regulating cardiac function [6]. Subsequent research led to the isolation of key active components, such as digoxin and digitoxin [7]. It was later discovered that CGs exert their cardiotonic effects through partial inhibition of the Na+/K+-ATPase (NKA) pump, resulting in increased intracellular calcium and enhanced myocardial contractility [8]. However, due to their narrow therapeutic window, many CGs have been replaced by safer alternatives—though digoxin remains in clinical use as the primary cardiotonic agent [9].
Despite their toxicity, CGs have regained attention for their selective cytotoxicity toward stressed or malignant cells, making them promising candidates for cancer therapy [10] and antiviral treatment [11]. Emerging evidence also suggests potential neuroprotective effects, further broadening their therapeutic relevance [12]. Mechanistically, NKA functions not only as an electrogenic pump that maintains ion homeostasis but also as a key membrane receptor involved in signal transduction [13]. CG binding induces conformational changes in NKA that activate signaling pathways such as Src/Ras/Raf/MEK/ERK, PI3K/Akt/mTOR, NF-κB, and PKC-dependent cascades [14]. These pathways regulate essential cellular functions including proliferation, differentiation, resistance to apoptosis, and inflammatory responses, implicating CGs in both physiological and pathological processes.
Recent research has substantially deepened our understanding of CGs. Novel types and producer species have been identified, and advanced techniques—such as molecular docking simulations and crystallography—have clarified the molecular basis of CG–NKA interactions. Additional molecular targets beyond NKA are also being explored. Efforts to overcome CGs’ narrow therapeutic index—such as the development of synthetic derivatives and targeted delivery methods—have further expanded their clinical potential. Moreover, co-evolutionary studies have illuminated the ecological drivers behind the structural diversity of CGs in plants.
This review provides a comprehensive examination of the historical significance and pharmacological activities of cardiac glycosides, explores recent molecular insights, and highlights their emerging roles in oncology, immunotherapy, antiviral therapy, and other fields.

2. Chemical Diversity and Origin of Cardiac Glycosides

The chemical structure of cardiac glycosides (CGs) comprises a steroidal aglycone core—specifically, a 5β,14β-androstane-3β,14-diol (Figure 1a)—linked to a sugar moiety at the C-3β position and a lactone ring at the C-17β position [15]. A defining feature of CGs is their characteristic “U”-shaped pharmacophore, formed by the cis–trans–cis fusion of the A/B, B/C, and C/D rings, respectively (Figure 1b). This conformation is essential for their interaction with Na+/K+-ATPase (NKA) and distinguishes them from other steroidal compounds [16]. While the steroid nucleus primarily confers pharmacological activity, the glycoside moiety modulates solubility, absorption, and overall bioavailability.
CGs are broadly classified into two major subtypes based on the structure of the lactone ring: cardenolides and bufadienolides. Cardenolides possess a five-membered α,β-unsaturated γ-lactone ring, whereas bufadienolides contain a six-membered α-pyrone ring [17]. This structural difference significantly influences their pharmacodynamics and toxicity. Bufadienolides typically exhibit higher potency and a narrower therapeutic index due to their stronger inhibition of NKA, which elevates the risk of cardiotoxicity [18]. In contrast, cardenolides—though still potent—generally present a more favorable therapeutic window and have been widely used in clinical settings to treat heart failure and atrial fibrillation [19]. Representative chemical structures of cardenolides and bufadienolides are depicted in Figure 1c and Figure 1d, respectively.

2.1. Cardenolides

Cardenolides are predominantly synthesized by plants as chemical defenses against herbivores. These compounds have been identified in over 17 plant families, encompassing hundreds of species across approximately 70–80 genera [20,21]. A recent review documented 295 cardenolides isolated from more than 30 higher plant species between 2010 and 2023 [22]. Among these, three novel cardenolides—castheveside A, castheveside B, and 3α-thevetiogenin—were recently isolated from Cascabela thevetia fruits [23].
The most prominent plant families and genera known to produce cardenolides include:
  • Apocynaceae: Asclepias, Nerium, Thevetia, Strophanthus, Cerbera, Calotropis (over 100 cardenolides identified).
  • Plantaginaceae: Digitalis.
  • Brassicaceae: Erysimum.
  • Ranunculaceae: Adonis.
  • Hyacinthaceae: Ornithogalum.
  • Moraceae: Antiaris.
  • Euphorbiaceae: Euphorbia.
  • Fabaceae: Corchorus.
Well-known cardenolide-producing species include Digitalis purpurea and Digitalis lanata (foxglove), Nerium oleander (oleander) [24,25], Thevetia peruviana (yellow oleander), Strophanthus gratus and S. kombe, Calotropis procera and C. gigantea (milkweeds) [26], Asclepias curassavica, Drimia maritima (Mediterranean squill), Convallaria majalis (lily of the valley), and Adonis vernalis (pheasant’s eye). Key cardenolides derived from these species include digoxin and digitoxin (Digitalis spp.), ouabain (Strophanthus spp.), convallatoxin (Convallaria majalis), calotropin and uscharin (Calotropis spp.), and thevetin (Thevetia peruviana). While these compounds have historically been employed to treat heart conditions, improper use can result in toxicity in both humans and animals.
Although plants are the primary producers of cardenolides, certain animal species are also capable of synthesizing these compounds. Notably, beetles in the family Chrysomelidae produce cardenolides such as sarmentogenin, periplogenin, and bipindogenin. Figure 2 (left) shows several plant and animal species that produce cardenolides, along with the names of the corresponding cardiac glycosides (CGs) they synthesize.

2.2. Bufadienolides

Bufadienolides are characterized by a six-membered α-pyrone (lactone) ring attached at the C-17β position of the steroidal (5β,14β-androstane-3β,14-diol) core [27]. The term “bufadienolide” originates from Bufo toads, whose venom contains these potent steroidal compounds. Historically, bufadienolide-containing plants were used as early as ancient Egypt, where squill (Drimia maritima, family Hyacinthaceae) was employed to treat heart disease [28]. The first bufadienolide glycoside to be structurally characterized was Scillaren A, identified in 1933 [29].
Beyond plants, bufadienolides are also produced by several animals, particularly toads of the Bufonidae family. The venom of approximately ten Bufo species has been confirmed to contain bufadienolides and their esters [30]. Traditional Asian medicines such as Ch’an Su and Senso are prepared from toad venom rich in these compounds [5]. Other amphibians, such as bufonid frogs of the genus Atelopus, produce related bufadienolides like telocinobufagin and bufotalin. Interestingly, lucibufagins—a structurally related subclass of bufadienolides—have also been identified in fireflies (Photinus ignitus and P. marginellus), where they serve as chemical deterrents against predators.
Among plants, bufadienolides have been reported in six families: Crassulaceae, Hyacinthaceae, Iridaceae, Melianthaceae, Ranunculaceae, and Santalaceae. The genus Kalanchoe (Hyacinthaceae) is particularly noteworthy for its high bufadienolide content [31]. In recent years, several studies have described the remarkable structural diversity of bufadienolides in Helleborus spp. [32,33], some of which exhibit potent cytotoxic effects against breast and cervical cancer cells [34].
Due to their cytotoxic and anticancer activities, bufadienolides are considered promising candidates for drug development, underscoring the need for further pharmacological and clinical investigation [35]. Among them, bufalin is one of the most extensively studied, having demonstrated anticancer [36], anti-inflammatory [37,38], and antiviral properties [39].
Figure 2 (right) depicts several bufadienolide-producing species, both animal and plant, along with the names of the corresponding cardiac glycosides (CGs) they synthesize.

2.3. Endogenous CGs

Although still debated, accumulating evidence suggests that mammals—including humans—endogenously produce compounds that are structurally and functionally analogous to cardiac glycosides (CGs). Due to their physiological relevance, these substances have been proposed as a novel class of steroid hormones [40]. A defining feature of CGs is their high-affinity binding to and inhibition of the sodium–potassium ATPase (NKA), which led researchers to hypothesize the existence of endogenous counterparts to plant- and animal-derived CGs [41]. This search was further motivated by efforts to identify a “hypothetical natriuretic hormone” involved in promoting sodium excretion and regulating blood pressure and vascular tone [42].
Between 1989 and 1991, a compound indistinguishable from ouabain—termed endogenous ouabain (EO)—was identified in human plasma [43]. EO is synthesized in the adrenal cortex, with its secretion regulated by epinephrine and angiotensin II [44]. Other endogenous cardiotonic steroids (ECTS), including EO, are also produced in the hypothalamus and are regulated by adrenocorticotropic hormone (ACTH), α-adrenergic and dopaminergic stimuli, angiotensin II (via AT2 receptor activation), hypoxia, and physical activity [40].
Elevated EO levels have been associated with hypertension, kidney dysfunction, and heart failure in both humans and animal models [45]. Additional endogenous CG-like compounds include digoxin, marinobufagenin [46], telocinobufagin [47], bufalin, 19-norbufalin, and proscillaridin A [46,47,48,49].
Despite their different origins, endogenous and exogenous CGs share several notable similarities, including a conserved steroidal structure, a lactone ring, and a high binding affinity for NKA. Both classes can inhibit the pump’s ion-transport function and initiate downstream signaling cascades. However, endogenous CGs are typically produced at nanomolar to picomolar concentrations and are thought to play physiological roles in maintaining sodium balance, vascular tone, and neuroendocrine regulation. In contrast, exogenous CGs—such as digoxin, ouabain, and bufalin—are administered at pharmacological doses and are primarily used to treat heart failure and arrhythmias.
Because both endogenous and exogenous CGs target overlapping NKA binding sites, competitive or synergistic interactions are possible. In pathological conditions characterized by elevated endogenous CG levels—such as heart failure or preeclampsia—these interactions may influence tissue responsiveness to therapeutic CGs, potentially enhancing efficacy or increasing toxicity. Therefore, understanding the interplay between endogenous and exogenous CGs is essential for optimizing treatment strategies and minimizing adverse effects, particularly in patients with altered endogenous CG profiles.

2.4. CG’s Derivatives

In recent years, several chemically modified derivatives of natural cardiac glycosides (CGs) have been developed to improve their physicochemical properties, selectivity, and therapeutic efficacy. Figure 3 illustrates the chemical structures of parent CGs and representative derivatives, which are described below.

2.4.1. Digoxin and Digitoxigenin Derivatives

Rocha and colleagues synthesized 21-Benzylidene Digoxin (21-BD) by introducing a styrene group into the lactone ring of digoxin—a modification shown to enhance biological activity [50]. Parreira et al. developed BD-15, a semi-synthetic γ-benzylidene derivative of digoxin that selectively enhances α3-NKA activity in the rat hippocampus and prefrontal cortex, with minimal impact on cardiac function [51]. Barathi et al. reported that DcB, a cyclobutyl derivative of digoxin, selectively inhibits the α2 isoform of NKA and effectively reduces intraocular pressure in ocular hypertensive nonhuman primates [52]. Additionally, O’Doherty’s group synthesized two digitoxigenin derivatives by modifying the sugar moiety with rhamnose or amicetose. Rhamnose substitution increased NKA affinity by 5–15-fold, while amicetose had no significant effect. Both derivatives elevated H2O2 levels, induced membrane lipid peroxidation, and reduced intracellular glutathione (GSH) levels [53].

2.4.2. Bufalin Derivatives

Lei et al. synthesized BF211, a bufalin derivative featuring a carbamate group at the C3 position. BF211 demonstrated stronger pro-apoptotic effects and lower toxicity compared to native bufalin [54]. Acetylation at the same position produced acetyl-bufalin, which exhibited enhanced antitumor activity against non-small-cell lung cancer [55]. Sampath and colleagues synthesized bufalin 2,3-ene and bufalin 3,4-ene, both of which retained the biological activity of bufalin while demonstrating reduced cytotoxicity [56].

2.4.3. Arenobufagin Derivatives

Chen and co-workers (2021) developed ZM226, a peptide-substituted arenobufagin derivative that showed enhanced antitumor activity and reduced cardiotoxicity [57]. Similarly, Tang et al. synthesized arenobufagin derivatives containing 3,11-bispeptide esters. Among these, ZM350 significantly inhibited tumor growth by 58.8% in an A549 nude mouse model, while maintaining low cardiac toxicity [58].
Overall, these chemically engineered CG derivatives demonstrate improved therapeutic indices, enhanced selectivity, and reduced systemic toxicity. Their development highlights the potential of CG analogs as next-generation therapeutic agents in both cancer and non-cancer applications.
Figure 3. Newly developed cardiac glycoside (CG) derivatives. Panels (ad) show the chemical structures of digoxin, digitoxin, bufalin, and arenobufagin, respectively, along with their corresponding derivatives. In each panel, the chemical structure of the parent CG is highlighted in violet.
Figure 3. Newly developed cardiac glycoside (CG) derivatives. Panels (ad) show the chemical structures of digoxin, digitoxin, bufalin, and arenobufagin, respectively, along with their corresponding derivatives. In each panel, the chemical structure of the parent CG is highlighted in violet.
Biomolecules 15 00885 g003

3. Na+/K+-ATPase, a Pump and a Receptor of CGs

Since its discovery by Jens Chr. Skou over five decades ago [59], Na+/K+-ATPase (NKA) has been closely associated with our understanding of cardiac glycoside (CG) pharmacology. Although both the conceptual framework of NKA function and CG pharmacology have evolved over time, their interconnection remains robust. Until recently, it was widely accepted that NKA was the sole molecular target of CGs and that all their known effects—both therapeutic and toxic—were mediated through this enzyme. While a few exceptions have emerged, this view remains largely valid. More recently, however, NKA has been recognized not only as an electrogenic ion pump but also as a membrane receptor. Upon binding with exogenous or endogenous CGs, NKA can activate diverse intracellular signaling pathways involved in both physiological regulation and pathogenesis.
Although several comprehensive reviews have discussed recent advances in the structural and functional understanding of NKA (e.g., Contreras et al., 2024 [13]), the following subsections provide a focused overview of its core characteristics, emphasizing how CGs influence NKA function—not only as an ion pump and signaling receptor but also in its lesser-known role as a cell adhesion molecule.

3.1. NKA Molecular Structure and Diversity

NKA is a heteromeric protein complex composed of three subunits: α, β, and γ (also known as FXYD) (Figure 4a,b). The α-subunit (~110 kDa) spans the membrane ten times and contains cytoplasmic N-, P-, and A-domains critical for ATP hydrolysis and ion transport. Its extracellular loops contribute to ion selectivity and interact with the β-subunit. Four α isoforms exist in mammals: α1 is ubiquitous and abundant in the kidney; α2 is expressed in muscle, heart, and brain; α3 is neuron-specific and essential for synaptic transmission; and α4 is testis-specific and crucial for sperm motility [60,61].
The β-subunit is a single-pass type II transmembrane protein with a short cytoplasmic N-terminus, a transmembrane domain, and a large extracellular region stabilized by disulfide bridges. It plays key roles in NKA assembly, membrane localization, and enzymatic stability [62]. Three β isoforms exist: β1 is ubiquitous; β2 (also known as AMOG) is enriched in glial and epithelial cells; and β3 is expressed in muscle, neurons, and other specialized tissues [63].
FXYD proteins are small, single-span transmembrane modulators that associate with the α-subunit to fine-tune pump activity in response to physiological factors such as ion concentrations, pH, and hormonal signals.

3.2. CG’s Influence on NKA Functions

3.2.1. CGs Inhibit NKA as a Pump

The primary role of NKA is to actively transport sodium (Na+) and potassium (K+) ions across the cell membrane against their concentration gradients, using energy from ATP hydrolysis. The α-subunit serves as the catalytic core, binding ATP, Na+, and K+, while the β-subunit ensures proper assembly and membrane localization. FXYD proteins dynamically regulate activity in response to metabolic and environmental cues.
CGs inhibit NKA by binding to a specific pocket on the extracellular domain of the α-subunit during the K+ binding and dephosphorylation stage of the pump cycle (Figure 4c,d). At this point, the pump adopts the E2-P conformation, having expelled Na+ and awaiting K+ binding. CG binding prevents dephosphorylation, locking the pump in an inactive state [64].
Recent cryo-electron microscopy (cryo-EM) and molecular docking studies have mapped the cardiotonic steroid (CTS) binding site in detail. Key interactions occur between CGs and extracellular loops and transmembrane domains (particularly M1–M6 and M9–M10). The steroid nucleus inserts into a hydrophobic cavity, the lactone ring at C-17 forms stabilizing interactions, and the sugar moiety at C-3β enhances affinity via hydrogen bonding [65]. The depth of steroid core insertion is critical for determining the mode and efficacy of inhibition [19,66].

3.2.2. NKA’s Role as a Signal-Transducing Receptor Activated by CG’s Binding

More than two decades ago, it was proposed that NKA could function as a receptor for ouabain and other CGs. This was later confirmed, especially for a subset of non-ion-transporting NKA molecules localized in caveolae—specialized membrane microdomains—where they form multiprotein complexes known as “signalosomes” [67]. Upon CG binding, NKA interacts with nearby signaling proteins, triggering intracellular pathways that regulate proliferation, differentiation, and apoptosis [68,69].
Figure 4. NKA, a pump and receptor of cardiac glycosides (CGs). (a) Topological profile of the constituent subunits. (b) Three-dimensional representation of the molecular surface of an NKA unit seen in profile, embedded in the membrane. (c) Same representation as in (b) but in molecular volume, to highlight the position occupied by a CG molecule. (d) Molecular surface view from the perspective indicated by the eye and arrow in scheme (c), highlighting the CG binding site. The images in (bd) were created in RCSB PDB (http://www.rcsb.org/) (accessed on 15 February 2025) [70] with the molecular graphics program Mol* [71] from PDB ID 7WYT [72].
Figure 4. NKA, a pump and receptor of cardiac glycosides (CGs). (a) Topological profile of the constituent subunits. (b) Three-dimensional representation of the molecular surface of an NKA unit seen in profile, embedded in the membrane. (c) Same representation as in (b) but in molecular volume, to highlight the position occupied by a CG molecule. (d) Molecular surface view from the perspective indicated by the eye and arrow in scheme (c), highlighting the CG binding site. The images in (bd) were created in RCSB PDB (http://www.rcsb.org/) (accessed on 15 February 2025) [70] with the molecular graphics program Mol* [71] from PDB ID 7WYT [72].
Biomolecules 15 00885 g004
The following subsections summarize the main CG-induced signaling pathways (see Figure 5).
Signaling Pathways Activated by CG Binding to NKA
Therapeutic doses of CGs can induce cardiac hypertrophy, suggesting a role in promoting growth beyond ion transport inhibition. Xie and Askari showed that ouabain induces immediate-early genes like c-fos, c-jun, and AP-1 in cardiac myocytes [73]. Ouabain activates Src kinase, which transactivates the epidermal growth factor receptor (EGFR), triggering the Ras/Raf/MEK/ERK cascade (Figure 5, yellow arrows) [74].
The partial inhibition of ouabain-induced ERK signaling by protein kinase C (PKC) inhibitors implicated additional pathways, including PLC/PKC signaling and Ca2+-dependent mechanisms (Figure 5, orange arrows) [75]. Moreover, ouabain enhances mitochondrial ROS production, activating MAPKs via Src-independent mechanisms [76,77]. CGs also stimulate PI3K/Akt/mTOR signaling independently of Src (Figure 5, blue arrows) [78].
Xie’s group showed that NKA, Src, and EGFR co-localize in caveolae and that NKA contains conserved caveolin-binding motifs, supporting its role as a scaffold in multiprotein signaling complexes [68].
CG-Induced Ca2+ Oscillations
In epithelial cells, ouabain induces intracellular calcium oscillations through a mechanism involving the inositol 1,4,5-trisphosphate receptor (IP3R) [79]. This activation does not rely on PLC-generated IP3 but rather on a direct interaction between NKA and IP3R within a complex stabilized by ankyrin B [77]. The LKK motif in the N-terminal domain of the NKA α-subunit is essential for IP3R binding [80].
This Ca2+ signaling promotes proliferation, adhesion, and apoptosis resistance via NF-κB activation. Phosphoproteomic analyses also reveal CAMK2G activation after ouabain stimulation [81].
CG-Induced ROS-Mediated Signaling
Reactive oxygen species (ROS) play a central role in CG-induced signaling through NKA. Molecules such as superoxide anion, hydrogen peroxide, and hydroxyl radicals act not only as metabolic by-products but also as secondary messengers in several NKA-mediated pathways [82]. Initial evidence linked ROS production to the activation of mitochondrial pathways downstream of Src activation (Figure 5, lime green path) [77]. Furthermore, antioxidant treatments were shown to partially inhibit ouabain-induced activation of MAPK, NF-κB, and protein synthesis, indicating that ROS are involved in both the catalytic and non-catalytic signaling effects of CGs [83].
Gene Regulation by CG-Induced Shifts in [Na+]i/[K+]i Ratios
CG-induced shifts in the intracellular sodium-to-potassium ratio can directly influence gene expression [84]. In endothelial cells, elevated extracellular Na+ modulates genes linked to vascular function [85]. Prolonged CG exposure alters hundreds of transcripts, as seen in HUVECs treated with ouabain or marinobufagenin [86]. One proposed mechanism involves monovalent cation sensors, such as DNA G-quadruplexes, that regulate transcription in response to ionic changes [87].

3.2.3. NKA’s Role in Cell–Cell Adhesion

Beyond its pumping and signaling roles, NKA contributes to intercellular adhesion in epithelial tissues [88]. In these cells, basolaterally localized β1 subunits from adjacent cells engage in direct interactions [89,90]. Fluorescence resonance energy transfer (FRET) studies confirm β1–β1 interactions, particularly at N-glycan-containing regions spanning residues 221–229 and 198–207 [91,92]. Structural alterations to β1 impair junction stability [93].
NKA also interacts with E-cadherin via the β1 subunit, reinforcing adherens junctions and epithelial cohesion [94]. In the retinal pigment epithelium (RPE), NKA localizes apically due to β2-mediated targeting [95]. Ouabain enhances β1–β1 adhesion via a Src-dependent pathway [96].
In the nervous system, the β2 subunit (AMOG) promotes neuron–glia adhesion [97,98]. Though heterologous expression suggests β2–β2 interactions, in vivo confirmation is pending [99]. Notably, astrocytes do not adhere to each other via AMOG, indicating that β2-mediated adhesion is selective for neuron–glia interactions [100].
These findings underscore NKA’s multifunctionality in epithelial and neuronal tissue architecture, influenced by β isoform composition and CG exposure.

4. Cardiac Glycosides: Modulators of Diverse Signaling Pathways

Recent advances reveal the remarkable diversity by which cardiac glycosides (CGs) modulate cellular signaling pathways. Their effects—either activating or inhibiting specific intracellular cascades—are highly dependent on cell type, tissue context, and physiological state. This complexity positions CGs as versatile modulators of critical biological processes such as proliferation, apoptosis, inflammation, and differentiation. These actions are shaped by context-dependent interactions within complex signaling networks.
The following subsections concisely overview major CG-influenced pathways, highlighting their biological significance and recent discoveries on specific CG regulation, offering insights into the therapeutic potential for cancer, fibrosis, and immune modulation.

4.1. PI3K/Akt Pathway

The phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) pathway is central to the regulation of cell growth, survival, metabolism, and proliferation. Dysregulation of this pathway is implicated in cancer, diabetes, and neurodegenerative diseases. Various CGs modulate PI3K/Akt signaling, affecting apoptosis, survival, and metastasis:
  • Cerberin inhibits PI3K/Akt/mTOR signaling in cancer [101].
  • Bufalin suppresses gastric cancer progression [102] and hepatoma invasion [14].
  • Strophanthidin and Lanatoside C attenuate PI3K/Akt/mTOR, reducing tumor growth and metastasis [103].
  • Oleandrin inhibits LRP4/MAPK/NF-κB, preventing osteoclast differentiation [104].

4.2. TGF-β/Smad Pathway

The transforming growth factor-beta (TGF-β)/Smad pathway governs cell proliferation, differentiation, and immune regulation. Its dysregulation contributes to cancer progression, fibrotic diseases, and immune disorders.
  • Digoxin inhibits TGF-β1/Smad signaling, preventing fibroblast differentiation into cancer-associated fibroblasts (CAFs) (CAFs) [105].
  • Periplocymarin activates TGF-β/Smad signaling, protecting against myocardial fibrosis [106].

4.3. HIF-1α Signaling

Hypoxia-inducible factor 1 (HIF-1), particularly its oxygen-sensitive subunit HIF-1α, orchestrates cellular responses to hypoxia by regulating genes involved in angiogenesis, metabolism, and survival [107]. Several CGs have been shown to inhibit HIF-1α signaling:
  • Digoxin, ouabain, and proscillaridin A inhibit HIF-1α protein synthesis and target gene expression [108].
  • Digoxin suppresses hypoxia-induced VEGF and NDRG1 expression [109].
  • Digitoxin inhibits HIF-1α and STAT3 in KRAS-mutant colon cancer [110].
  • Bufalin enhances photodynamic therapy by inhibiting SRC-3/HIF-1α [110] and targets mTOR/HIF-1α in ovarian carcinoma [111].
  • Cardenolides from Calotropis gigantea inhibit HIF-1 transcriptional activity [112].

4.4. JAK/STAT Pathway

The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway mediates responses to cytokines and growth factors, playing essential roles in cell proliferation, immune regulation, and inflammation. Several CGs modulate this pathway in disease-specific contexts:
  • Periplogenin inhibits JAK2/3-STAT3 signaling to reduce synovial proliferation in arthritis [113].
  • Bufalin suppresses JAK/STAT to reduce inflammation in cancer and cardiovascular diseases [38].
  • Periplocymarin alleviates cardiac hypertrophy via JAK2/STAT3 inhibition [114].
  • Bufothionine induces autophagy in hepatoma-bearing mice through JAK2/STAT3 blockade [115].
  • Convallatoxin inhibits colorectal cancer proliferation via JAK2/STAT3 and mTOR/STAT3 pathways [116].
  • Peruvoside also targets PI3K/Akt/mTOR in cancer cells [117].

4.5. PERK/elF2α/ATF4/CHOP Pathway

This signaling axis is a major component of the unfolded protein response (UPR) triggered by endoplasmic reticulum (ER) stress, determining cell fate through adaptation or apoptosis.
  • Oleandrin activates the PERK/eIF2α/ATF4/CHOP pathway, inducing immunogenic death in breast cancer cells [118].
  • Neriifolin induces ER stress-mediated apoptosis in prostate cancer by activating PERK and CHOP, impairing DNA repair mechanisms [119].
As demonstrated, CGs exhibit complex and context-dependent modulation of key signaling pathways—including PI3K/Akt, TGF-β/Smad, HIF-1α, JAK/STAT, and UPR—across diverse pathological states. This multifaceted interaction underpins their significant therapeutic potential in cancer (inhibiting growth, survival, metastasis), fibrosis (modulating TGF-β), inflammation (targeting NF-κB, JAK/STAT), and immune regulation. Future research should focus on elucidating the precise context-specific mechanisms of individual CGs to harness their full potential for targeted therapies.

5. Cardiac Glycosides in Physiological and Pathological Processes

Initially studied for their cardiotonic and toxic properties, CGs are now recognized for their broader biological relevance. As illustrated in Figure 6, emerging research highlights their involvement in a wide range of physiological and pathological processes with significant biomedical implications.
From a physiological perspective, the discovery that certain CGs are endogenously synthesized has stimulated interest in their diverse biological roles [120]. CGs function as modulators of the epithelial phenotype and play critical roles in regulating blood pressure, maintaining volume homeostasis, and controlling plasma sodium levels.
Pathologically, CGs influence numerous molecular mechanisms implicated in major diseases. Cancer has emerged as a particularly important area, with increasing evidence supporting the impact of CGs on tumor progression and therapeutic response. Additionally, CGs have shown potential as anti-inflammatory and antiviral agents, as well as modulators of immune and nervous system-related disorders—further underscoring their broad therapeutic relevance [20,121,122].
The following subsections provide an updated overview of the roles of various CGs in these key physiological and pathological contexts.

5.1. CGs’ Role in Cardiac Function and Regulation

CGs are best known for their effects on cardiac function. Although hundreds of CGs are found in nature, only a few—ouabain, digitoxin, and digoxin—have been used clinically. For nearly two centuries, these compounds were employed to manage heart conditions. However, ouabain and digitoxin were eventually discontinued due to their narrow therapeutic windows and the emergence of safer alternatives, such as beta-blockers and ACE inhibitors [123,124]. Currently, digoxin remains the only CG in clinical use, primarily for treating heart failure with reduced ejection fraction (HFrEF) and atrial fibrillation [125,126].
Digoxin’s relatively short half-life allows for precise dosing and easier toxicity management. Standard dosages range from 0.125 to 0.25 mg/day, with adjustments required in patients with renal impairment [9,127,128]. In overdose scenarios, digoxin-specific antibody fragments (Digibind) are used as an antidote to neutralize circulating digoxin and promote its renal elimination [129].
CGs exert positive inotropic effects by inhibiting NKA in cardiac myocytes. This inhibition raises intracellular sodium, reduces sodium–calcium exchange, and increases intracellular calcium—particularly in the sarcoplasmic reticulum. The resulting calcium surge during depolarization enhances myocardial contractility, improving cardiac output and relieving heart failure symptoms. Additionally, CGs exhibit vagomimetic effects, slowing heart rate and atrioventricular conduction, which is beneficial in atrial fibrillation. Nonetheless, due to their narrow therapeutic index, CGs require careful monitoring to prevent potentially fatal arrhythmias.

5.2. CGs as Modulators of Salt (Sodium) and Blood Pressure

Over the past three decades, research has demonstrated that both endogenous and exogenous CGs influence cardiovascular physiology beyond direct cardiac effects, particularly in fluid balance, sodium regulation, and blood pressure control [120,121].
The short-term administration of exogenous CGs—including cardenolides (e.g., ouabain, G-strophanthin, ouabagenin, dihydroouabain) and bufadienolides (e.g., marinobufagenin, telocinobufagin, proscillaridin A)—can enhance cardiac contractility, elevate blood pressure, and promote natriuresis at the renal tubular level [130]. However, long-term exposure to certain CGs is associated with cardiac and renal hypertrophy and the development of hypertension [131]. Interestingly, digoxin does not induce hypertension with chronic use and may even counteract ouabain-induced hypertensive effects [132].
Endogenous cardiac glycosides (ECGs)—such as endogenous ouabain (EO) and marinobufagenin (MBG)—are crucial regulators of blood pressure and sodium balance [133]. Their plasma levels fluctuate with sodium intake, systemic blood pressure, stress, and physical activity. Elevated ECG concentrations are observed in pathological conditions associated with sodium dysregulation, including hypertension [134], hyperaldosteronism [135], renal artery stenosis [136], preeclampsia [45], and heart failure.
ECGs also interact with each other and key systems regulating sodium homeostasis, such as the renin–angiotensin–aldosterone system (RAAS) [137]. EO levels inversely correlate with cardiac function in heart failure [121]. ECGs are implicated in the pathogenesis of several hypertensive conditions [138], including those linked to chronic kidney disease and salt sensitivity [139]. Remarkably, rostafuroxin (PST2238), a digitoxigenin derivative, mitigates EO- and ouabain-induced hypertension.
In summary, ECGs play critical roles in sodium and fluid balance. Elevated sodium in the midbrain stimulates hippocampal ouabain release, triggering ACTH and angiotensin II secretion. These hormones promote adrenal synthesis and the release of ECGs from cholesterol precursors [140].

5.3. CGs as Modulators of the Epithelial Phenotype

Epithelia are a primordial structural form in Metazoa, serving as barriers and regulators of solute exchange between compartments. Epithelial cells are characterized by apical-basolateral polarity and specialized junctions, including tight junctions, adherens junctions, desmosomes, and gap junctions [141,142].
The initial observation that ouabain induces the detachment of MDCK cells from substrates and each other led Cereijido and colleagues to hypothesize that CGs modulate epithelial phenotype [143,144]. Subsequent evidence confirms that nanomolar concentrations of ouabain modulate key epithelial structures:
  • Tight junctions [145].
  • Adherens junctions [146].
  • Gap junctions [147,148,149].
Other CGs, including digoxin and marinobufagenin, similarly enhance gap junctional intercellular communication [150].
Beyond junctional effects, CGs influence epithelial polarization. For example, ouabain accelerates primary cilium formation in confluent MDCK cells—a marker of epithelial maturity [151].
CGs also regulate ion channel expression, including voltage-gated potassium channels [152] and TRPV4 channels [153]. Notably, these effects depend on cell–cell contact, suggesting that CG signaling is context-sensitive.
Additionally, ouabain stimulates prostaglandin E2 (PGE2) production and release, which further enhances gap junction communication [154], reinforcing its role in epithelial connectivity through both direct and indirect mechanisms.
Mechanistically, these effects originate from CG binding to Na+/K+-ATPase, which functions as both an ion pump and a signal transducer. This interaction activates key intracellular pathways, including c-Src, ERK1/2, PI3K, and Rho/ROCK, thereby orchestrating the phenotypic changes observed in epithelial architecture [155]. Figure 7 schematically illustrates the signaling cascades and epithelial structures modulated by CGs through NKA binding.

5.4. CGs’ Influence in Cancer Processes

Cancer comprises a group of over 100 diseases characterized by uncontrolled cell proliferation and the ability to invade surrounding tissues and metastasize to distant organs. While common types include breast, lung, and colorectal cancers, others—such as pancreatic cancer, glioblastoma, and hepatocellular carcinoma—are particularly aggressive and treatment-resistant [156]. Tumor progression involves several hallmark biological capabilities, including self-sufficiency in growth signals, resistance to growth inhibition, the evasion of apoptosis, limitless replicative potential, sustained angiogenesis, and metastatic dissemination [157]. These features result from complex molecular alterations, notably in transcription factors (TFs) that govern inflammation, hypoxia response, proliferation, epithelial–mesenchymal transition (EMT), and cellular plasticity. Key TFs include NF-κB, HIF-1, c-Myc, AP-1, and STAT3 [158].
The potential anticancer activity of cardiac glycosides (CGs) was first suggested by Stenkvist et al. in 1979, who observed reduced tumor aggressiveness in breast cancer patients receiving CG treatment [159]. Since then, numerous studies have reported the anticancer effects of various CGs across diverse malignancies, including breast, lung, pancreatic, colorectal, and liver cancers [160]. These effects encompass multiple cancer hallmarks, including the induction of programmed cell death, the inhibition of metastasis, the suppression of angiogenesis, and the disruption of cell cycle progression. Additionally, CGs modulate epigenetic mechanisms and contribute to overcoming drug resistance, highlighting their potential as multifaceted anticancer agents [161]. Table 1 summarizes recent findings on the anticancer properties of various CGs and their effects on key signaling pathways across different cancer types.

5.4.1. CGs as Inducers of Cancer Cell Death

A defining feature of CG-mediated anticancer activity is their ability to selectively trigger cell death in tumor cells through diverse mechanisms. Below, we review their roles in various forms of programmed cell death, emphasizing recent studies and context-specific effects.
CGS as Senolytics
Cellular senescence is a state of stable, irreversible cell cycle arrest triggered by stressors such as DNA damage, oxidative stress, or oncogene activation. Although senescent cells cease proliferating, they remain metabolically active and secrete a pro-inflammatory milieu termed the senescence-associated secretory phenotype (SASP) [199]. In cancer, senescence plays a dual role: it can prevent tumor progression by arresting damaged cells but also promote tumorigenesis by fostering chronic inflammation and remodeling the tumor microenvironment. This paradox has spurred interest in senolytics—agents that selectively eliminate senescent cells—as therapeutic tools in oncology and age-related diseases [200].
Several CGs have emerged as potent senolytic compounds (reviewed in [201]). In a high-throughput screen, Triana-Martínez et al. identified proscillaridin A, ouabain, digoxin, bufalin, cinobufagin, peruvoside, digitoxin, and convallatoxin as promising senolytics [202]. Guerrero et al. further demonstrated that CGs such as ouabain, bufalin, ouabagenin, k-strophanthin, and strophanthidin selectively induce apoptosis in senescent cells via diverse mechanisms [203]. More recently, machine learning-based screens identified periplocin and oleandrin as additional CG-derived senolytics [204]. Resibufogenin, a bufadienolide from toad venom, was shown to eliminate senescent cells via a caspase-3-dependent apoptotic mechanism [205].
CGs as Inducers of Apoptosis
Apoptosis is a tightly regulated form of programmed cell death critical for development and tissue homeostasis. It proceeds via two primary pathways: the intrinsic (mitochondrial) pathway, triggered by internal stressors and mediated by cytochrome c, Apaf-1, and caspase-9; and the extrinsic (death receptor) pathway, initiated by ligands such as FasL or TNF-α, leading to caspase-8 activation. Both pathways converge on effector caspases (e.g., caspase-3), which execute cellular dismantling. These pathways are tightly controlled by regulatory proteins, including the Bcl-2 family and inhibitor of apoptosis proteins (IAPs). In cancer, the dysregulation of apoptosis via p53 mutation, Bcl-2 overexpression, or caspase inhibition contributes to tumor persistence and therapy resistance [206,207,208].
Numerous CGs—including digoxin and ouabain—induce apoptosis in cancer cells, primarily by inhibiting NKA pumping activity, which leads to elevated intracellular calcium and reactive oxygen species (ROS) generation. These changes activate both intrinsic and extrinsic apoptotic pathways, disrupt mitochondrial function, modulate apoptotic regulators such as Bcl-2 and p53, and activate caspases. CGs also inhibit pro-survival pathways such as NF-κB, enhancing apoptotic signaling. Importantly, many CGs exhibit cancer-selective toxicity, offering therapeutic advantages [160,209,210]. Table 2 summarizes CG-induced apoptotic mechanisms across various cancer models.
In addition to apoptosis, several non-apoptotic forms of regulated cell death play crucial roles in cancer biology and therapy. Recent studies have highlighted the capacity of cardiac glycosides (CGs) to modulate these pathways—particularly ferroptosis, pyroptosis, and parthanatos—offering novel therapeutic strategies, especially against resistant or heterogeneous tumors.
CGs as Inducers of Ferroptosis
Ferroptosis is a distinct form of regulated cell death driven by iron-dependent lipid peroxidation and oxidative damage, characterized by mitochondrial shrinkage and membrane rupture [230,231,232,233]. Key regulators include glutathione peroxidase 4 (GPX4), the cystine/glutamate antiporter system (SLC7A11), and antioxidant signaling via Nrf2. Several bufadienolides—including arenobufagin, bufotalin, bufalin, and resibufogenin—have been shown to induce ferroptosis in cancer cells.
Arenobufagin induces ferroptosis in glioblastoma and gastric cancer by promoting ROS accumulation and suppressing the Nrf2 pathway [163,234,235]. Bufalin causes lipid peroxidation and downregulates the SLC7A11/GPX4 axis in breast cancer [236]. Bufotalin inhibits GPX4 and induces ferroptosis in lung and colorectal cancer via nanoparticle delivery systems [236] Resibufogenin activates ferroptosis in lung cancer through a non-coding RNA-mediated signaling axis involving transferrin receptor regulation [195].
CGs as Inducers of Pyroptosis
Pyroptosis is a pro-inflammatory form of regulated cell death characterized by cell swelling, membrane rupture, and the release of cytokines such as interleukin-1β (IL-1β) and IL-18. It is mediated by inflammasome assembly, caspase-1 activation, and gasdermin D (GSDMD) cleavage [206,237]. CGs have been identified as inducers of pyroptosis in cancer and non-cancer contexts. For instance, ouabain activates the NLRP3 inflammasome and induces pyroptosis in cardiomyocytes and immune cells [238]. More recently, bufalin-loaded CaCO3 nanoparticles (BCNPs@gel) have been shown to induce Ca2+ overload-mediated pyroptosis in tumor models, underscoring the therapeutic potential of CG-based nanomedicines [239].
CGs as Inducers of Parthanatos
Parthanatos is a caspase-independent cell death pathway triggered by hyperactivation of poly(ADP-ribose) polymerase-1 (PARP-1), leading to NAD+ depletion, mitochondrial dysfunction, and eventual cell death [240]. The synthetic cardenolide ZINC253504760 has been reported to induce parthanatos in leukemia cells [241]. Additionally, hellebrigenin promotes PARP-1-mediated parthanatos in oral squamous cell carcinoma [223], further supporting the role of CGs in engaging this emerging form of regulated cell death.
CGs as Inducers of Autophagic Cell Death
Autophagic cell death (ACD) is a regulated form of cell death characterized by excessive autophagy, resulting in the self-digestion and degradation of essential cellular components. Unlike apoptosis, ACD occurs independently of caspase activation and is marked by autophagosome accumulation and lysosomal degradation. It is mediated by autophagy-related proteins such as Beclin-1 and LC3 [242,243]. While physiologically important in development and immunity, ACD plays dual roles in cancer—either suppressing tumor growth or enabling survival under metabolic stress [242,244].
Cardiac glycosides, including cardenolides (e.g., digoxin, digitoxin, ouabain, oleandrin, lanatoside C, Anvirzel and bufadienolides (e.g., bufalin, proscillaridin A), modulate autophagy in a context-dependent manner [210]. They can either stimulate or inhibit autophagy. Inhibition is often associated with autosis, an NKA-dependent form of autophagic cell death. Conversely, CGs can promote autophagy through mechanisms involving AMPK/mTOR and Src/MEK/ERK pathway activation, ROS production, mitochondrial stress, TFEB nuclear translocation, and JNK signaling.
CG-induced autophagy has therapeutic potential, with varying outcomes. In lung cancer, it enhances treatment efficacy, while in gastric and liver cancers, it sensitizes cells to autophagy inhibitors—supporting its value in combinatorial strategies [245].
CGs as Inducers of Immunogenic Cell Death
Immunogenic cell death (ICD) is a regulated form of cell death that activates adaptive antitumor immunity, primarily through the stimulation of cytotoxic T lymphocytes. ICD is characterized by the release of damage-associated molecular patterns (DAMPs) such as calreticulin (CRT), ATP, high-mobility group box 1 (HMGB1), and type I interferons, which promote dendritic cell activation and immune cell recruitment [246,247,248]. These features make ICD a promising strategy in cancer immunotherapy [249].
CGs such as digoxin, digitoxin, ouabain, and lanatoside C have been shown to induce the hallmark features of ICD, with immune-stimulating effects comparable to chemotherapeutics like methotrexate [250]. Other CGs, including Scillaren A, proscillaridin, and digitoxigenin, also elicit ICD responses [251]. Notably, digoxin enhances CRT exposure and ATP release when used in combination with cisplatin(IV)–polymer conjugates [252]. In breast cancer models, oleandrin promotes ICD through CRT exposure and the release of HMGB1, HSP70/90, and ATP [118], further supporting the immunomodulatory capacity of CGs.

5.4.2. CGs as Inducers of Cell Cycle (G2/M) Arrest

The G2/M checkpoint ensures that cells do not enter mitosis until DNA replication is complete and genomic integrity is preserved. The dysfunction of this checkpoint is common in cancer and contributes to uncontrolled proliferation. Agents that induce G2/M arrest can exploit this vulnerability to inhibit tumor growth, promote DNA damage responses, and trigger apoptosis [253].
Cardiac glycosides have been shown to induce G2/M arrest in various cancer types:
  • Lanatoside C in human prostate cancer cells [184].
  • Bufalin in head and neck cancer [254] and lung cancer [255].
  • Telocinobufagin in head and neck squamous carcinoma [256], oral squamous carcinoma [223], and HL-60 leukemia cells [257].
  • Arenobufagin in A549 lung cancer cells [211].
  • ZINC253504760 in leukemia [241]
  • Resibufogenin in glioma [258].
  • Cinobufagin in colorectal, hepatocellular, and melanoma cells [220].
  • Digoxin in lung cancer cells [259].
These findings suggest that CG-induced G2/M arrest is a broadly conserved and therapeutically exploitable mechanism.

5.4.3. CGs as Modulators of Angiogenesis

Hypoxia in tumors—caused by rapid proliferation and poor vascularization—triggers the stabilization of hypoxia-inducible factor 1α (HIF-1α), a transcription factor that induces pro-survival and pro-angiogenic genes, including vascular endothelial growth factor (VEGF) [260,261,262]. Targeting hypoxia and angiogenesis is a major strategy in cancer therapy [263].
Several CGs inhibit angiogenesis by blocking HIF-1α synthesis, even under hypoxic conditions [264]. For example:
  • Digitoxin reduces HIF-1α and STAT3 expression, induces apoptosis, and limits proliferation in KRAS-mutant colon cancer cells [222]. It also impairs glioma stemness by downregulating HIF-1α [109].
  • Bufalin exerts potent antiangiogenic effects by inhibiting multiple signaling axes, including STAT3, mTOR/HIF-1α, and VEGF, across various malignancies such as ovarian and liver cancers [111,265,266].
  • CG derivatives from Calotropis gigantea exhibit greater inhibitory effects on HIF-1α than digoxin, indicating the potential for enhanced antiangiogenic efficacy [112].
Moreover, advanced drug delivery systems—such as hypoxia-responsive micelles and dual-loaded nanocarriers—have improved CG efficacy against HIF-1α, augmenting their therapeutic potential [267,268].

5.4.4. CGs as Inhibitors of EMT and Metastasis

Metastasis—the leading cause of cancer mortality—is driven by epithelial–mesenchymal transition (EMT), where epithelial cells acquire mesenchymal features that enhance motility and invasiveness. EMT is mediated by transcription factors like Snail, Slug, and Twist and is associated with poor prognosis and resistance to therapy [269,270].
Multiple CGs have demonstrated anti-EMT activity via diverse mechanisms:
  • Arenobufagin suppresses EMT by:
    • Downregulating c-MYC/Nrf2 in colorectal cancer [271].
    • Inhibiting IKKβ/NF-κB in lung cancer [166].
    • Reducing β-catenin levels in prostate cancer [164].
  • Bufalin exerts multi-pathway EMT inhibition:
    • Blocking the c-Kit/Slug axis [272].
    • Inhibiting STAT3 signaling [273].
    • Modulating the SRC-3/c-Myc pathway in colorectal cancer [168].
    • Targeting PI3K/AKT/mTOR in gastric cancer [102].
    • Suppressing Src signaling in non-small cell lung cancer [255].
  • Bufotalin inhibits EMT through the STAT3/EMT axis in triple-negative breast cancer [217].
  • Cinobufagin:
    • Induces Enkurin expression and regulates the β-catenin/c-Jun/MYH9/p53 axis in nasopharyngeal carcinoma [274].
    • Inhibits FAK/STAT3 signaling in triple-negative breast cancer [275].
  • Cinobufotalin suppresses EMT in hepatocellular carcinoma by downregulating β-catenin [276].
  • Telocinobufagin shows promising EMT-inhibitory activity in undifferentiated thyroid carcinoma, although the specific molecular mechanisms remain to be clarified [196].
Together, these studies highlight the potential of CGs to suppress EMT and limit metastatic spread.
In summary, cardiac glycosides (CGs) have emerged as potent modulators of cancer biology, acting across multiple hallmarks of malignancy. In addition to their classical cardiotonic effects, CGs exhibit anticancer properties through diverse mechanisms, including the induction of apoptosis and alternative forms of regulated cell death (e.g., ferroptosis, pyroptosis, parthanatos, autophagic death, immunogenic death). They also modulate cell cycle progression, suppress angiogenesis, and inhibit EMT and metastasis. Their selective cytotoxicity toward cancer and senescent cells, coupled with their immunomodulatory potential, positions CGs as promising candidates for oncological therapeutics. Future studies should aim to define optimal dosing, delivery strategies, and combination regimens to maximize their efficacy while minimizing toxicity.

5.5. Cardiac Glycosides as Immunomodulators: Dual Roles in Inflammation and Adaptive Immunity

Emerging evidence indicates that cardiac glycosides (CGs) possess immunomodulatory properties, acting through a range of molecular targets and signaling pathways that regulate immune cell activation, cytokine production, and inflammatory responses. Among these, digoxin has garnered particular attention due to its interaction with the nuclear receptors retinoic acid receptor-related orphan receptors γ and γt (RORγ and RORγt)—transcription factors central to the differentiation of T helper 17 (Th17) cells and the regulation of pro-inflammatory cytokines. In addition to modulating RORγ signaling, CGs influence key intracellular pathways, including NF-κB, PI3K/Akt, and JAK/STAT, highlighting their potential therapeutic utility in autoimmune diseases, chronic inflammatory disorders, and cancer-associated immune dysregulation [210,277].

5.5.1. Digoxin as an Inhibitor of Th17 Cell Differentiation

Pioneering work by Huh et al. [278] identified digoxin as a selective RORγt antagonist using a Drosophila-based reporter assay. Digoxin inhibited Th17 cell differentiation and suppressed IL-17 transcription without significantly affecting other T cell subsets. In murine models of experimental autoimmune encephalomyelitis (EAE), digoxin treatment delayed disease onset and mitigated neurological symptoms. Furthermore, synthetic derivatives such as 20,22-dihydrodigoxin-21,23-diol and digoxin-21-salicylidene demonstrated low toxicity while selectively inhibiting IL-17 production in human CD4+ T cells. Complementary studies by Fujita-Sato et al. confirmed that digoxin directly binds to the ligand-binding domain of RORγt, effectively suppressing Th17 differentiation while sparing Th1 cells and RORα signaling [279].
The anti-inflammatory effects of digoxin have been validated across various disease models:
  • Arthritis: Digoxin reduced inflammation and disease progression in both adjuvant-induced [280] and collagen-induced arthritis [281].
  • Multiple sclerosis: In EAE models, digoxin attenuated disease severity by inhibiting RORγt and promoting oligodendrocyte differentiation and remyelination. Notably, full remission was achieved when digoxin was combined with tolerance-inducing nanoparticle therapy [282]
  • Colitis: Digoxin alleviated colitis symptoms by downregulating IL-17A and IL-17F expression while upregulating the anti-inflammatory cytokine IL-10 in a manner independent of TNF-α signaling [283].
  • Rheumatoid arthritis (RA): Digoxin suppressed Th17 differentiation and reduced the production of key inflammatory cytokines (IL-1β, IL-6, IL-17, IL-23) without significantly altering Th1-related markers [284].

5.5.2. Contradictory Findings: CGs as RORγ Agonists

Despite initial findings characterizing CGs as RORγ antagonists, subsequent studies have revealed agonist-like activity under specific conditions, challenging their previously assumed selectivity. Using a HepG2 cell-based reporter system, Karaś et al. identified multiple CGs—including strophanthidin, digoxigenin, and dihydroouabain—as RORγ and RORγt agonists that enhanced IL-17A and IL-17F expression in human Th17 cells [285]. Strikingly, even nanomolar concentrations of digoxin elicited comparable agonist-like responses.
Further expanding on this, Karwaciak et al. tested a panel of 16 CGs (e.g., bufalin, oleandrin, lanatoside C, digitoxin) and observed the upregulation of RORγ target genes such as G6PC, as well as the increased expression of pro-inflammatory cytokines (IL-17A, IL-17F, IFN-γ) and chemokines (CXCL10) in primary human Th17 cells [286]. Supporting the potential pro-inflammatory effects of CGs, a population-based cohort study found a statistically significant association between long-term digoxin use and the increased incidence of psoriasis, particularly among patients with comorbidities such as heart failure, diabetes, and hyperlipidemia [287].
These contrasting observations likely reflect dose-dependent, context-specific, and cell type-dependent effects of CGs, influenced by factors such as immune microenvironment, metabolic state, and the experimental system used.

5.5.3. RORγ-Independent Immunomodulation

Beyond RORγ signaling, several CGs exert immunomodulatory effects through alternative pathways, affecting diverse immune cell populations and inflammatory circuits:
  • Periplogenin inhibited the JAK2/3–STAT3 axis, reduced pro-inflammatory cytokine production, and suppressed synoviocyte proliferation and migration in models of rheumatoid arthritis [113].
  • Cinobufagin (CBG) enhanced the release of IL-1β and TNF-α, partially through the activation of the PI3K/Akt/mTOR pathway [288].
  • Bufalin, identified through systems biology approaches, modulated the expression of immune-related genes including S100B, BIRC5, MMP9, and EGFR, suggesting immunoregulatory roles in breast cancer progression [289].
  • Gamabufotalin selectively reduced regulatory T cell (Treg) populations while sparing peripheral blood mononuclear cells, indicating a potential role in enhancing antitumor immune responses [290].
  • Convallatoxin (CNT) attenuated vascular inflammation in atherosclerosis by promoting anti-inflammatory M2 macrophage polarization via the PPARγ–Integrin αvβ5 pathway; this effect was reversed by the pharmacological inhibition of PPARγ [291].
These findings reveal that CGs can modulate a broad range of immune responses—including T cell differentiation, cytokine secretion, macrophage activation, and tissue inflammation—independently of RORγ activity.
In summary, CGs exhibit a dualistic and context-dependent immunomodulatory profile, acting through both RORγ-dependent and RORγ-independent mechanisms. While some CGs suppress Th17-driven inflammation by antagonizing RORγt, others may paradoxically enhance pro-inflammatory responses via RORγ activation. Additionally, several CGs influence alternative pathways such as JAK/STAT, PI3K/Akt, and PPARγ, impacting innate and adaptive immunity. These complex and sometimes contradictory effects underscore the need for a nuanced understanding of dosage, chemical structure, target cell type, and disease context when considering CGs for immunotherapeutic applications. Future research should aim to delineate the molecular determinants that govern these divergent immune outcomes, thereby enabling the rational development of CG-based therapies for autoimmune diseases, inflammatory disorders, and cancer immunotherapy.

5.6. Cardiac Glycosides as Antiviral Agents

Cardiac glycosides (CGs) have emerged as promising broad-spectrum antiviral agents, exhibiting activity against a wide variety of DNA and RNA viruses. Compounds such as digoxin, lanatoside C, bufalin, ouabain, digitoxin, convallatoxin, proscillaridin A, oleandrin, gitoxin, deslanoside, and k-strophanthidin have demonstrated efficacy against numerous pathogens, including herpes simplex virus type 1 (HSV-1), human cytomegalovirus (HCMV), human immunodeficiency virus (HIV), Zika virus (ZIKV), dengue virus (DENV), influenza A virus (IAV), Ebola virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), infectious hematopoietic necrosis virus (IHNV), and vaccinia virus [161,292].

5.6.1. Mechanisms of Antiviral Action

The antiviral effects of CGs are primarily mediated through their interaction with NKA, particularly the α1 subunit. The binding of CGs to NKA alters intracellular ion homeostasis—most notably by reducing intracellular K+ levels—which disrupts essential processes in the viral life cycle, such as RNA synthesis, protein translation, and post-transcriptional processing. This mechanism has been well-documented in infections caused by RSV and IAV [293].
Beyond ionic disruption, CGs also activate NKA-dependent signaling cascades, including the Src–EGFR–Ras–Raf–MEK–ERK pathway. The activation of this pathway has been shown to block clathrin-mediated endocytosis, a key entry mechanism for RSV, coronaviruses, and other enveloped viruses [294]. In HIV, CGs interfere with the RNA splicing machinery, leading to over-splicing and the subsequent depletion of functional viral mRNAs [295]. In HSV-1 infections, CGs reduce viral egress, resulting in lower extracellular viral titers [295]. Importantly, CGs act post-attachment, primarily affecting intracellular stages of viral replication and assembly [296].

5.6.2. Experimental Evidence Supporting Antiviral Activity

Numerous experimental studies provide strong support for the antiviral potential of CGs:
  • Coronaviruses: Digitoxin and ouabain strongly inhibited human coronaviruses HCoV-229E, HCoV-OC43, and SARS-CoV-2 in primary human nasal epithelial cells and lung organoids. The observed antiviral effects were associated with the activation of the MEK and JNK signaling pathways [297].
  • Bunyamwera virus: Digoxin inhibited viral replication in Vero cells by reducing viral protein synthesis and altering cell cycle progression. These effects were abolished in cells expressing a digoxin-resistant NKA, confirming the role of NKA inhibition in mediating antiviral activity [298].
  • IHNV: Bufalin suppressed both viral attachment and RNA replication in vitro and significantly improved survival and reduced viral burden in infected rainbow trout in vivo. The mechanism was linked to NKA modulation [39].
  • HSV-1: Lanatoside C inhibited HSV-1 replication by activating the NRF2 pathway. NRF2 nuclear translocation reduced viral gene expression and preserved nerve fiber integrity in vivo, highlighting NRF2 as a potential therapeutic target [299].
  • SARS-CoV-2 (in silico studies): Computational docking studies by Qayed et al. demonstrated that ouabain, digitoxin, digoxin, and proscillaridin bind strongly to key viral targets, including PLpro, Mpro, RNA-dependent RNA polymerase (RdRp), and AAK1. Ouabain was identified as a dual inhibitor of PLpro and Mpro, while digitoxin specifically targeted RdRp [300].
  • Zika virus: Ouabain inhibited ZIKV replication in human neural stem and progenitor cells. In a murine model of congenital Zika syndrome, it significantly reduced viral loads in fetal tissues, enhanced neurogenesis, mitigated fetal growth restriction, and decreased levels of pro-inflammatory cytokines [301].
In summary, CGs represent a novel class of host-targeted antiviral agents with diverse and potent effects against both RNA and DNA viruses. Through NKA inhibition, the disruption of ion homeostasis, and the activation of downstream signaling pathways, CGs interfere with viral entry, replication, RNA splicing, and protein translation. Their broad efficacy against multiple viral families—ranging from flaviviruses and herpesviruses to retroviruses and coronaviruses—underscores their therapeutic potential.
Importantly, their ability to modulate host pathways rather than targeting viral components directly may reduce the likelihood of resistance development. Nonetheless, given their narrow therapeutic window, dose-dependent toxicity, and cell-type specificity, further mechanistic studies, structure–activity relationship analyses, and clinical trials are essential to optimize their use and assess safety in antiviral therapy.

5.7. Cardiac Glycosides as Neuromodulators: Emerging Roles in the Nervous System

Beyond their well-characterized cardiac and anticancer properties, cardiac glycosides (CGs) are increasingly recognized as modulators of central nervous system (CNS) function. Recent research indicates that CGs can regulate neuroinflammation, synaptic plasticity, and cognitive processes, suggesting their potential for repurposing in neurological and psychiatric disorders.

5.7.1. Alzheimer’s Disease (AD)

In preclinical models of AD, CGs—particularly ouabain and digoxin—have demonstrated neuroprotective effects:
  • Ouabain improved cognitive performance in transgenic AD mice by promoting anti-inflammatory microglial polarization through TREM2 upregulation and PI3K/Akt pathway activation [302]. It also reduced tau pathology by activating TFEB and autophagy [303].
  • Digoxin enhanced memory and neuronal survival in a rat model of sporadic AD by suppressing TNF-α and restoring choline acetyltransferase (ChAT) activity [304].
  • CGs have also been shown to upregulate miR-132, a neuroprotective microRNA typically downregulated in AD. This upregulation was associated with reduced tau expression and the preservation of neuronal integrity [305].

5.7.2. Bipolar Disorder (BD)

The exogenous administration of ouabain in rodent models induced manic- and depression-like behaviors, recapitulating cognitive and affective symptoms characteristic of BD. These effects were accompanied by elevated levels of pro-inflammatory cytokines and impaired BDNF/TrkB signaling, a pathway critical for synaptic plasticity and mood regulation [306,307].
Interestingly, a hypothesized link between COVID-19-related adrenal damage and reduced endogenous ouabain production has been proposed to contribute to BD symptom exacerbation, pointing to a potential neuroendocrine role of CGs in mood regulation [308].

5.7.3. Epilepsy and Multiple Sclerosis

  • In a chronic epilepsy (kindling) model, digoxin enhanced the efficacy of sodium valproate, improved seizure control, and reduced markers of neuroinflammation, supporting its use as a potential adjuvant therapy [309].
  • In models of demyelination, digoxin promoted oligodendrocyte differentiation and myelin repair, particularly when combined with antigen-specific immune tolerance, showing promise for the treatment of multiple sclerosis (MS) [282].

5.7.4. Synaptic Function and Excitotoxicity

CGs also modulate synaptic signaling and protect against excitotoxic damage:
  • Ouabain prevented NMDA-induced excitotoxicity by stabilizing the interaction of Na+/K+-ATPase (NKA) with NCX and NMDARs within lipid rafts, thereby regulating calcium influx and maintaining synaptic integrity [310].
  • Digoxin facilitated dendritic spine formation and improved motor learning, particularly in mice with deficits in the neurotrypsin–agrin signaling pathway [311].

5.7.5. Cognitive Effects in Vascular and Cardiac Contexts

Cognitive benefits of CGs have also been observed in cardiovascular and cerebrovascular disease models:
  • In elderly patients with heart failure, digoxin administration was associated with improved cognitive performance [312].
  • In a mouse model of chronic cerebral hypoperfusion, digoxin restored glymphatic function and reduced white matter injury, an effect dependent on aquaporin-4 activity [313].
Collectively, these findings highlight the emerging potential of cardiac glycosides as modulators of CNS function. Their ability to influence neuroinflammation, neuroprotection, synaptic signaling, and cognitive outcomes provides a strong rationale for further investigation into their use in neurodegenerative, neuropsychiatric, and neurovascular disorders.

6. Cardiac Glycosides Target Molecules Beyond Na+/K+-ATPase

While the NKA remains the canonical receptor mediating the effects of CGs, growing evidence reveals that CGs can also interact directly with a variety of non-NKA targets, including kinases, membrane receptors, transcription factors, and epigenetic regulators. These interactions highlight the multifaceted mechanisms by which CGs exert their biological effects.

6.1. Kinases and Signaling Proteins

Several CGs directly inhibit intracellular kinases that regulate oncogenic and inflammatory pathways:
  • CAMKK2 is inhibited by bufalin, leading to the suppression of intrahepatic cholangiocarcinoma via the inhibition of the Wnt/β-catenin pathway [171].
  • JAK1, a key mediator of cytokine signaling, is also directly targeted by bufalin, which disrupts the JAK1–ACAP4 interaction, thereby blocking IL-6-induced downstream signaling [314].
  • CDK9 and STAT3 have been implicated as targets of acetyl-bufalin in non-small cell lung cancer, contributing to its potent anti-tumor effects [55].

6.2. Receptor Tyrosine Kinases

The bufadienolide resibufogenin (RBF) binds directly to the ATP-binding site of VEGFR2, inhibiting its phosphorylation and downstream angiogenic signaling. This impairs endothelial function and suppresses tumor vascularization, especially in triple-negative breast cancer [194].

6.3. MAPK Pathway and Transcriptional Regulators

Transcriptomic and molecular docking analyses suggest that CGs can directly modulate MAPK signaling:
  • In MCF-7 breast cancer cells, lanatoside C, peruvoside, and strophanthidin were predicted to target MAPK1 and EGR1 [315].
  • In hepatocellular carcinoma, bufalin was shown to interact with MAPK1, MAPK14, PRKCA, EIF4E, and APEX1, with binding validated by docking studies and Western blotting [266].

6.4. Nuclear Receptors and Transcriptional Regulators

CGs have also been shown to target transcriptional regulators involved in immune and oncogenic signaling:
  • CGs bind to the ligand-binding domain of RORγ/RORγT, nuclear receptors involved in immune regulation and tumor progression [286].
  • SRC-3, a transcriptional coactivator of c-Myc, is a direct target of bufalin in chemoresistant colorectal cancer. Bufalin-mediated downregulation of SRC-3 suppresses c-Myc expression and metastasis. The overexpression of either SRC-3 or c-Myc reverses these effects, confirming a functional dependency [168].

6.5. Other Membrane Receptors

The membrane receptor LRP4 is targeted by both oleandrin and digoxin. Oleandrin inhibits osteoclastogenesis via LRP4 in bone models [104], while digoxin exerts anti-inflammatory and chondroprotective effects through LRP4 in nucleus pulposus cells and osteoarthritis models [316,317].

6.6. RNA-Binding Proteins

Telocinobufagin (TBG) binds to LARP1, a translational regulator in the mTOR signaling pathway. TBG disrupts the LARP1–mTOR interaction, leading to the inhibition of metastasis in undifferentiated thyroid cancer [196].

6.7. Oncoproteins and Apoptosis Regulators

Bufalin targets the BFAR (Bifunctional Apoptosis Regulator), an anti-apoptotic protein that activates the PI3K/Akt/mTOR pathway. Bufalin binding downregulates the BFAR and suppresses downstream oncogenic signaling in gastric cancer [102].

6.8. Epigenetic Regulators

The cardiac glycoside AT2, produced by Antiaris toxicaria, directly inhibits UHRF1, a chromatin regulator involved in DNA methylation and gene silencing, thereby revealing a novel NKA-independent mechanism of action [318].

6.9. Hypoxia Response Regulators

In a pharmacological screening, HIF-1α was identified as a direct target of digoxin, which inhibited its protein synthesis and transcriptional activity. The anti-tumor effects of digoxin were reversed in cells overexpressing HIF-1α, confirming functional relevance [108].
In summary, beyond NKA, cardiac glycosides interact with a broad spectrum of molecular targets, including kinases, nuclear receptors, RNA-binding proteins, and epigenetic regulators. These diverse interactions significantly contribute to the anticancer, anti-inflammatory, and immunomodulatory effects of CGs, reinforcing their potential for therapeutic repurposing across multiple disease contexts.

7. Discussion

Cardiac glycosides (CGs), originally developed for the treatment of heart failure and arrhythmias, have garnered renewed interest as potential therapeutic agents across a wide spectrum of non-cardiac diseases, including cancer, viral infections, autoimmune disorders, and neurodegenerative conditions. This paradigm shift is supported by growing evidence that CGs modulate diverse cellular pathways, extending far beyond their classical role as Na+/K+-ATPase (NKA) inhibitors.
Although the inhibition of NKA remains a fundamental mechanism underlying the effects of CGs, recent studies have uncovered their capacity to modulate multiple intracellular signaling pathways, including NF-κB, MAPK, PI3K/Akt, JAK/STAT, and mTOR. These pathways regulate key cellular processes such as inflammation, proliferation, apoptosis, and immune responses. Importantly, CGs have been shown to trigger a range of regulated cell death modalities—including apoptosis, ferroptosis, pyroptosis, parthanatos, autophagic cell death, and immunogenic cell death—which may be exploited to overcome therapy resistance in refractory malignancies.
Beyond their well-characterized role in modulating NKA, CGs have been shown to directly engage a wide variety of non-NKA molecular targets. These include receptor tyrosine kinases (e.g., VEGFR2), transcriptional regulators (e.g., SRC-3, RORγ), epigenetic enzymes (e.g., UHRF1), and RNA-binding proteins (e.g., LARP1). These alternative interactions help to explain the context-dependent effects of CGs across different tissues and disease models, broadening their mechanistic and therapeutic repertoire.
In the field of virology, CGs interfere with multiple stages of the viral life cycle—including entry, replication, and RNA processing—particularly in infections caused by coronaviruses, herpesviruses, HIV, and flaviviruses. These antiviral effects are primarily mediated through the disruption of host ion homeostasis and the inhibition of key signaling pathways such as Src–EGFR–Ras–MEK–ERK, highlighting their promise as host-targeted antiviral agents with a lower likelihood of inducing viral resistance.
In immunology, CGs—most notably digoxin—exert immunomodulatory effects by inhibiting RORγt, a nuclear receptor crucial for Th17 cell differentiation. These effects have demonstrated therapeutic potential in models of autoimmune encephalomyelitis, arthritis, and colitis. However, contrasting findings have also emerged: some CGs, or digoxin at specific concentrations or in certain cellular contexts, have shown agonistic activity on RORγ, promoting IL-17 expression. These context- and structure-specific dualities emphasize the complexity of CG pharmacology and underscore the need for further mechanistic clarification.
From a neurological perspective, CGs display neuroprotective properties in preclinical models of Alzheimer’s disease, epilepsy, multiple sclerosis, and vascular cognitive impairment. These effects are attributed to their ability to suppress neuroinflammation, enhance autophagy, regulate calcium signaling, and promote synaptic plasticity. Moreover, CGs have been implicated in mood disorders such as bipolar disorder, with both therapeutic benefits and pathophysiological contributions suggested, depending on endogenous or exogenous exposure.
Despite this broad therapeutic promise, the narrow therapeutic index of CGs—characterized by a fine line between effective and toxic doses—remains a major challenge to clinical translation. Cardiotoxicity, in particular, limits their systemic use and necessitates careful dosing and monitoring.
To overcome these limitations, two principal strategies are under active investigation:
  • Medicinal chemistry approaches to develop CG derivatives with reduced toxicity and enhanced selectivity.
  • Advanced delivery systems, such as nanoparticles, micelles, and conjugates, which enable tissue-specific delivery and reduce systemic exposure.
Together, these innovations seek to expand the therapeutic window of CGs, facilitating their repurposing as viable treatments for complex modern diseases such as cancer, autoimmune conditions, and neurodegeneration.

8. Conclusions

Cardiac glycosides (CGs), long regarded solely as cardiotonic agents, have emerged as a diverse class of pharmacological modulators with compelling potential across oncology, virology, immunology, and neurology. Their capacity to engage both canonical targets—such as Na+/K+-ATPase—and non-canonical effectors, including kinases, nuclear receptors, transcriptional co-regulators, and epigenetic modulators, situates CGs at the crossroads of multiple therapeutic domains.
As the landscape of CG research evolves, key challenges must be systematically addressed. These include improving target selectivity, enhancing therapeutic safety, and developing advanced delivery platforms capable of minimizing off-target toxicity. In parallel, a deeper mechanistic understanding of CG actions—particularly their context-dependent effects and interactions with cellular signaling networks—remains essential for their rational clinical repurposing.
Moving forward, a multidisciplinary strategy that integrates systems biology, structural pharmacology, and nanotechnology will be vital to fully exploit the therapeutic potential of this ancient yet highly adaptable class of compounds. With continued innovation and rigorous investigation, cardiac glycosides may be repositioned as modern therapeutics for some of the most pressing and complex diseases of our time.

Author Contributions

Conceptualization, A.P., R.G.C. and C.F.-M.; methodology, A.P. and C.F.-M.; software, A.P.; validation, A.P.; investigation, A.P. and C.F.-M.; resources, A.P. and R.G.C.; writing—original draft preparation, A.P. and C.F.-M.; writing—review and editing, A.P., R.G.C. and C.F.-M.; supervision, A.P. and R.G.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received funding from the National Research Council of Mexico (CONAHCYT) CF-2019-7357.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ACTHAdrenocorticotropic hormone
ACDAutophagic cell death
AMOGAdhesion Molecule on Glia
AP-1Activator protein 1
ATPAdenosine triphosphate
CAMKCalcium/calmodulin-dependent protein kinase
CAFsCancer-associated fibroblasts
CaMKKβCalcium/calmodulin-dependent protein kinase kinase β
CGsCardiac glycosides
CRTCalreticulin
DAMPsDamage-associated molecular patterns
EGFREpidermal growth factor receptor
EMTEpithelial–mesenchymal transition
EOEndogenous ouabain
EREndoplasmic reticulum
ERKExtracellular signal-regulated kinase
FXYDSingle-span transmembrane regulatory protein family of NKA
GPX4Glutathione peroxidase 4
GSHGlutathione
HCMVHuman cytomegalovirus
HIF-1αHypoxia-inducible factor 1 alpha
HMGB1High mobility group box 1
HSVHerpes simplex virus
ICDImmunogenic cell death
IFN-γInterferon gamma
ILInterleukin
IP3RInositol 1,4,5-trisphosphate receptor
JAKJanus kinase
MAPKMitogen-activated protein kinase
mTORMammalian target of rapamycin
NKANa+/K+-ATPase
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NLRP3NOD-, LRR-, and pyrin domain-containing protein 3
NRF2Nuclear factor erythroid 2–related factor 2
PARP-1Poly(ADP-ribose) polymerase 1
PERKPKR-like ER kinase
PI3KPhosphoinositide 3-kinase
PKCProtein kinase C
PLCPhospholipase C
RORγRetinoic acid-related orphan receptor gamma
ROSReactive oxygen species
SASPSenescence-associated secretory phenotype
SrcProto-oncogene tyrosine-protein kinase Src
STATSignal transducer and activator of transcription
TFEBTranscription factor EB
Th17T helper 17 cells
TGF-βTransforming growth factor beta
TNF-αTumor necrosis factor alpha
TRAILTNF-related apoptosis-inducing ligand
UPRUnfolded protein response
VEGFVascular endothelial growth factor

References

  1. Radford, D.J.; Gillies, A.D.; Hinds, J.A.; Duffy, P. Naturally Occurring Cardiac Glycosides. Med. J. Aust. 1986, 144, 540–544. [Google Scholar] [CrossRef] [PubMed]
  2. Stoll, A. Les glycosides cardiotoniques [Cardiotonic glycosides]. Praxis 1948, 37, 156. [Google Scholar] [PubMed]
  3. Badri, S.; Pavitra, P.; Sameena, D.; Anitha, G.; Asifa, S.K.; Mahesh, K.; Pushpalatha, E. A Review on Medicinal Plants Containing Glycosides. UPI J. Pharm. Med. Health Sci. 2023, 6, 8–15. [Google Scholar] [CrossRef]
  4. Jia, J.; Li, J.; Zheng, Q.; Li, D. A Research Update on the Antitumor Effects of Active Components of Chi Nese Medicine ChanSu. Front. Oncol. 2022, 12, 1014637. [Google Scholar] [CrossRef]
  5. Zou, D.; Wang, Q.; Chen, T.; Sang, D.; Yang, T.; Wang, Y.; Gao, M.; He, F.; Li, Y.; He, L.; et al. Bufadienolides Originated from Toad Source and Their Anti-Inflammatory Activity. Front. Pharmacol. 2022, 13. [Google Scholar] [CrossRef]
  6. Withering, W. An Account of the Foxglove, and Some of Its Medical Uses; Cambridge University Press: Cambridge, UK, 2014; ISBN 1-108-07586-X. [Google Scholar]
  7. Tantivatana, P.; Wright, S.E. Estimation of Digoxin and Digitoxin in Digitalis Lanata. J. Pharm. Pharmacol. 1958, 10, 189–193. [Google Scholar] [CrossRef]
  8. Skou, J.C. The Influence of Some Cations on an Adenosine Triphosphatase from Peripheral Nerves. Biochim. Biophys. Acta 1957, 23, 394–401. [Google Scholar] [CrossRef]
  9. Crane, A.D.; Militello, M.; Faulx, M.D. Digoxin Is Still Useful, but Is Still Causing Toxicity. Cleve. Clin. J. Med. 2024, 91, 489–499. [Google Scholar] [CrossRef]
  10. Škubník, J.; Pavlíčková, V.; Rimpelová, S. Cardiac Glycosides as Immune System Modulators. Biomolecules 2021, 11, 659. [Google Scholar] [CrossRef]
  11. Newman, R.A.; Sastry, K.J.; Arav-Boger, R.; Cai, H.; Matos, R.; Harrod, R. Antiviral Effects of Oleandrin. J. Exp. Pharmacol. 2020, 12, 503–515. [Google Scholar] [CrossRef]
  12. Fender, J.; Klöcker, J.; Boivin-Jahns, V.; Ravens, U.; Jahns, R.; Lorenz, K. “Cardiac Glycosides”-Quo Vaditis?-Past, Present, and Future? Naunyn. Schmiedebergs Arch. Pharmacol. 2024, 397, 9521–9531. [Google Scholar] [CrossRef] [PubMed]
  13. Contreras, R.G.; Torres-Carrillo, A.; Flores-Maldonado, C.; Shoshani, L.; Ponce, A. Na(+)/K(+)-ATPase: More than an Electrogenic Pump. Int. J. Mol. Sci. 2024, 25, 6122. [Google Scholar] [CrossRef] [PubMed]
  14. Sheng, X.; Zhu, P.; Zhao, Y.; Zhang, J.; Li, H.; Zhao, H.; Qin, J. Effect of PI3K/AKT/mTOR Signaling Pathway on Regulating and Controlling the Anti-Invasion and Metastasis of Hepatoma Cells by Bufalin. Recent Pat. Anticancer. Drug Discov. 2021, 16, 54–65. [Google Scholar] [CrossRef] [PubMed]
  15. Bartnik, M.; Facey, P. Chapter 7—Glycosides. In Pharmacognosy, 2nd ed.; McCreath, S.B., Clement, Y.N., Eds.; Academic Press: Cambridge, MA, USA, 2024; pp. 103–165. ISBN 978-0-443-18657-8. [Google Scholar]
  16. Chapelon, A.-S.; Moraléda, D.; Rodriguez, R.; Ollivier, C.; Santelli, M. Enantioselective Synthesis of Steroids. Tetrahedron 2007, 63, 11511–11616. [Google Scholar] [CrossRef]
  17. Joubert, J.P.J. Cardiac Glycosides. In Toxicants of Plant Origin; CRC Press: Boca Raton, FL, USA, 1989; ISBN 978-1-003-41827-6. [Google Scholar]
  18. Barrueto, F.J.; Kirrane, B.M.; Cotter, B.W.; Hoffman, R.S.; Nelson, L.S. Cardioactive Steroid Poisoning: A Comparison of Plant- and Animal-Derived Compounds. J. Med. Toxicol. Off. J. Am. Coll. Med. Toxicol. 2006, 2, 152–155. [Google Scholar] [CrossRef]
  19. Azalim, P.; do Monte, F.M.; Rendeiro, M.M.; Liu, X.; O’Doherty, G.A.; Fontes, C.F.; Leitão, S.G.; Quintas, L.E.M.; Noël, F. Conformational States of the Pig Kidney Na(+)/K(+)-ATPase Differently Affect Bufadienolides and Cardenolides: A Directed Structure-Activity and Structure-Kinetics Study. Biochem. Pharmacol. 2020, 171, 113679. [Google Scholar] [CrossRef]
  20. Ren, J.; Gao, X.; Guo, X.; Wang, N.; Wang, X. Research Progress in Pharmacological Activities and Applications of Cardiotonic Steroids. Front. Pharmacol. 2022, 13, 902459. [Google Scholar] [CrossRef]
  21. Kowsalya, K.; Vidya, N.; Halka, J.; Preetha, J.S.Y.; Saradhadevi, M.; Sahayarayan, J.J.; Gurusaravanan, P.; Arun, M. Plant Glycosides and Glycosidases: Classification, Sources, and Therapeutic Insights in Current Medicine. Glycoconj. J. 2025, 42, 107–124. [Google Scholar] [CrossRef]
  22. He, Y.-L.; Yang, H.-Y.; Zhang, L.; Gong, Z.; Li, G.-L.; Gao, K. Research Progress on Plant-Derived Cardenolides (2010–2023). Chem. Biodivers. 2024, 21, e202401460. [Google Scholar] [CrossRef]
  23. Long, J.; Ouyang, J.-C.; Luo, Y.-H.; Wu, Q.-J.; Liao, X.-T.; Chen, Z.-L.; Wang, Q.-L.; Liang, X.-Y.; Liu, L.; Yang, X.-M.; et al. Three New Cardenolides from the Fruits of Cascabela Thevetia (L.) Lippold and Their Cytotoxic Activities. Nat. Prod. Res. 2024, 38, 211–219. [Google Scholar] [CrossRef]
  24. Mishra, V.K.; Rathour, B.K.; Mishra, S.K.; Sagar, R. Cardenolide and Pregnatriene Compounds from the Roots of Nerium Oleander. Nat. Prod. Res. 2021, 35, 4177–4181. [Google Scholar] [CrossRef] [PubMed]
  25. Sharma, R.; Singh, S.; Tewari, N.; Dey, P. A Toxic Shrub Turned Therapeutic: The Dichotomy of Nerium Oleander Bioactivities. Toxicon Off. J. Int. Soc. Toxinology 2023, 224, 107047. [Google Scholar] [CrossRef] [PubMed]
  26. Rajkovic, J.; Novakovic, R.; Grujic-Milanovic, J.; Ydyrys, A.; Ablaikhanova, N.; Calina, D.; Sharifi-Rad, J.; Al-Omari, B. An Updated Pharmacological Insight into Calotropin as a Potential Therapeutic Agent in Cancer. Front. Pharmacol. 2023, 14, 1160616. [Google Scholar] [CrossRef] [PubMed]
  27. Ji Ram, V.; Goel, A.; Pratap, R. Chapter 3 - Chemistry of Isolated 2-Pyranones. In Isolated Pyranones; Ji Ram, V., Goel, A., Pratap, R., Eds.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 11–175. ISBN 978-0-12-821216-5. [Google Scholar]
  28. Krenn, L.; Kopp, B. Bufadienolides from Animal and Plant Sources. Phytochemistry 1998, 48, 1–29. [Google Scholar] [CrossRef]
  29. Stoll, A.; Suter, E.; Kreis, W.; Bussemaker, B.B.; Hofmann, A. Die Herzaktiven Substanzen Der Meerzwiebel. Scillaren A. Helv. Chim. Acta 1933, 16, 703–733. [Google Scholar] [CrossRef]
  30. Asrorov, A.M.; Kayumov, M.; Mukhamedov, N.; Yashinov, A.; Mirakhmetova, Z.; Huang, Y.; Yili, A.; Aisa, H.A.; Tashmukhamedov, M.; Salikhov, S.; et al. Toad Venom Bufadienolides and Bufotoxins: An Updated Review. Drug Dev. Res. 2023, 84, 815–838. [Google Scholar] [CrossRef]
  31. Stefanowicz-Hajduk, J.; Hering, A.; Gucwa, M.; Hałasa, R.; Soluch, A.; Kowalczyk, M.; Stochmal, A.; Ochocka, R. Biological Activities of Leaf Extracts from Selected Kalanchoe Species and Their Relationship with Bufadienolides Content. Pharm. Biol. 2020, 58, 732–740. [Google Scholar] [CrossRef]
  32. Guo, J.; Chen, F.; Zhang, W.; Bai, H.; Li, L.; Ma, Y.; Yang, Z. Separation of Bufadienolides from Helleborus Thibetanus Franch. by a Combination Approach Involving Macroporous Resin Column Chromatography and Gradient Countercurrent Chromatography. J. Sep. Sci. 2024, 47, e2400145. [Google Scholar] [CrossRef]
  33. Yokosuka, A.; Inomata, M.; Yoshizawa, Y.; Iguchi, T.; Mimaki, Y. Bufadienolides and Ecdysteroids from the Whole Plants of Helleborus Niger and Their Cytotoxicity. J. Nat. Med. 2021, 75, 393–402. [Google Scholar] [CrossRef]
  34. Potterat, O.; Kaufmann, M.; Tschopp, C.; Caj, M.; Reinhardt, J.K.; Prescimone, A.; Shah, D.; Baumgartner, S.; Sciotti, M.-A.; Suter-Dick, L. Bufadienolides from Helleborus Foetidus and Their Cytotoxic Properties on MCF-7 Breast Cancer Cells. Phytochemistry 2025, 230, 114329. [Google Scholar] [CrossRef]
  35. Deng, L.-J.; Li, Y.; Qi, M.; Liu, J.-S.; Wang, S.; Hu, L.-J.; Lei, Y.-H.; Jiang, R.-W.; Chen, W.-M.; Qi, Q.; et al. Molecular Mechanisms of Bufadienolides and Their Novel Strategies for Cancer Treatment. Eur. J. Pharmacol. 2020, 887, 173379. [Google Scholar] [CrossRef] [PubMed]
  36. Tang, D.; Feng, Y.; Lu, J.; Jia, L.; Shen, D.; Shang, J.; Chen, T.; Yin, P.; Chen, J.; Wang, J. Global Trends in Bufalin Application Research for Cancer from 2003 to 2022: A Bibliometric and Visualised Analysis. Heliyon 2024, 10, e24395. [Google Scholar] [CrossRef] [PubMed]
  37. Niu, X.; Sun, W.; Tang, X.; Chen, J.; Zheng, H.; Yang, G.; Yao, G. Bufalin Alleviates Inflammatory Response and Oxidative Stress in Experimental Severe Acute Pancreatitis through Activating Keap1-Nrf2/HO-1 and Inhibiting NF-κB Pathways. Int. Immunopharmacol. 2024, 142, 113113. [Google Scholar] [CrossRef] [PubMed]
  38. Kaur, G.; Devi, S.; Sharma, A.; Sood, P. Pharmacological Insights and Role of Bufalin (Bufadienolides) in Inflammation Modulation: A Narrative Review. Inflammopharmacology 2024, 32, 3057–3077. [Google Scholar] [CrossRef]
  39. Zhao, J.-Z.; Xu, L.-M.; Li, L.-F.; Ren, G.-M.; Shao, Y.-Z.; Liu, Q.; Lu, T.-Y. Traditional Chinese Medicine Bufalin Inhibits Infectious Hematopoietic Necrosis Virus Infection in Vitro and in Vivo. Microbiol. Spectr. 2024, 12, e0501622. [Google Scholar] [CrossRef]
  40. Schoner, W. Endogenous Cardiac Glycosides, a New Class of Steroid Hormones. Eur. J. Biochem. 2002, 269, 2440–2448. [Google Scholar] [CrossRef]
  41. Blaustein, M.P.; Hamlyn, J.M. Sensational Site: The Sodium Pump Ouabain-Binding Site and Its Ligands. Am. J. Physiol. Cell Physiol. 2024, 326, C1120–C1177. [Google Scholar] [CrossRef]
  42. Hamlyn, J.M.; Blaustein, M.P. Endogenous Ouabain: Recent Advances and Controversies. Hypertension 2016, 68, 526–532. [Google Scholar] [CrossRef]
  43. Hamlyn, J.M.; Blaustein, M.P.; Bova, S.; DuCharme, D.W.; Harris, D.W.; Mandel, F.; Mathews, W.R.; Ludens, J.H. Identification and Characterization of a Ouabain-like Compound from Human Plasma. Proc. Natl. Acad. Sci. USA 1991, 88, 6259–6263. [Google Scholar] [CrossRef]
  44. Manunta, P.; Messaggio, E.; Casamassima, N.; Gatti, G.; Carpini, S.D.; Zagato, L.; Hamlyn, J.M. Endogenous Ouabain in Renal Na(+) Handling and Related Diseases. Biochim. Biophys. Acta 2010, 1802, 1214–1218. [Google Scholar] [CrossRef]
  45. Socha, M.W.; Chmielewski, J.; Pietrus, M.; Wartęga, M. Endogenous Digitalis-like Factors as a Key Molecule in the Pathophysiology of Pregnancy-Induced Hypertension and a Potential Therapeutic Target in Preeclampsia. Int. J. Mol. Sci. 2023, 24, 12743. [Google Scholar] [CrossRef] [PubMed]
  46. 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] [PubMed]
  47. Komiyama, Y.; Dong, X.H.; Nishimura, N.; Masaki, H.; Yoshika, M.; Masuda, M.; Takahashi, H. A Novel Endogenous Digitalis, Telocinobufagin, Exhibits Elevated Plasma Levels in Patients with Terminal Renal Failure. Clin. Biochem. 2005, 38, 36–45. [Google Scholar] [CrossRef] [PubMed]
  48. Goto, A.; Ishiguro, T.; Yamada, K.; Ishii, M.; Yoshioka, M.; Eguchi, C.; Shimora, M.; Sugimoto, T. Isolation of a Urinary Digitalis-like Factor Indistinguishable from Digoxin. Biochem. Biophys. Res. Commun. 1990, 173, 1093–1101. [Google Scholar] [CrossRef]
  49. Oda, M.; Kurosawa, M.; Numazawa, S.; Tanaka, S.; Akizawa, T.; Ito, K.; Maeda, M.; Yoshida, T. Determination of Bufalin-like Immunoreactivity in Serum of Humans and Rats by Time-Resolved Fluoroimmunoassay for Using a Monoclonal Antibody. Life Sci. 2001, 68, 1107–1117. [Google Scholar] [CrossRef]
  50. Pessôa, M.T.C.; Valadares, J.M.M.; Rocha, S.C.; Silva, S.C.; McDermott, J.P.; Sánchez, G.; Varotti, F.P.; Scavone, C.; Ribeiro, R.I.M.A.; Villar, J.A.F.P.; et al. 21-Benzylidene Digoxin Decreases Proliferation by Inhibiting the EGFR/ERK Signaling Pathway and Induces Apoptosis in HeLa Cells. Steroids 2020, 155, 108551. [Google Scholar] [CrossRef]
  51. Parreira, G.M.; Faria, J.A.; Marques, S.M.S.; Garcia, I.J.P.; Silva, I.F.; De Carvalho, L.E.D.; Villar, J.A.F.P.; Machado, M.V.; de Castro Lima, M.; Barbosa, L.A.; et al. The γ-Benzylidene Digoxin Derivative BD-15 Increases the A3-Na, K-ATPase Activity in Rat Hippocampus and Prefrontal Cortex and No Change on Heart. J. Membr. Biol. 2021, 254, 189–199. [Google Scholar] [CrossRef]
  52. Barathi, V.A.; Katz, A.; Chaudhary, S.; Li, H.-L.; Tal, D.M.; Marcovich, A.; Do, C.-W.; Karlish, S.J.D. A Digoxin Derivative That Potently Reduces Intraocular Pressure: Efficacy and Mechanism of Action in Different Animal Models. Am. J. Physiol. Cell Physiol. 2024, 326, C1505–C1519. [Google Scholar] [CrossRef]
  53. Valadares, J.M.M.; Azalim-Neto, P.; Liu, X.; Carrozza, N.C.; O’Doherty, G.A.; Quintas, L.E.M.; Barbosa, L.A. Pharmacodynamic Characterization and Evaluation of Oxidative Stress Effects of Digitoxigenin Derivatives on HeLa Cells. J. Membr. Biol. 2025. [Google Scholar] [CrossRef]
  54. Lei, M.; Xiao, Z.; Ma, B.; Chen, Y.; Liu, M.; Liu, J.; Guo, D.; Liu, X.; Hu, L. Synthesis and Biological Evaluation of Bufalin-3-Yl Nitrogen-Containing-Carbamate Derivatives as Anticancer Agents. Steroids 2016, 108, 56–60. [Google Scholar] [CrossRef]
  55. Yang, L.; Zhou, F.; Zhuang, Y.; Liu, Y.; Xu, L.; Zhao, H.; Xiang, Y.; Dai, X.; Liu, Z.; Huang, X.; et al. Acetyl-Bufalin Shows Potent Efficacy against Non-Small-Cell Lung Cancer by Targeting the CDK9/STAT3 Signalling Pathway. Br. J. Cancer 2021, 124, 645–657. [Google Scholar] [CrossRef] [PubMed]
  56. Sampath, V.; Horesh, N.; Sasi, B.; Zannadeh, H.; Pogodin, I.; Singh, S.V.; Deutsch, J.; Lichtstein, D. Synthesis and Biological Evaluation of Novel Bufalin Derivatives. Int. J. Mol. Sci. 2022, 23, 4007. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, B.; Wang, C.; Ma, J.; Ma, H.; Wang, Y.; Zhang, H.; Zhu, Y.; Yao, J.; Luo, C.; Miao, Z.; et al. Discovery of 3-Peptide Substituted Arenobufagin Derivatives as Potent Antitumor Agents with Low Cardiotoxicity. Steroids 2021, 166, 108772. [Google Scholar] [CrossRef] [PubMed]
  58. Tang, W.; Zhang, Y.; Yang, K.; Ma, J.; Dong, L.; Wu, C.; Lv, R.; Wang, C.; Luo, C.; Zhang, H.; et al. Discovery of Novel 3,11-Bispeptide Ester Arenobufagin Derivatives with Potential in Vivo Antitumor Activity and Reduced Cardiotoxicity. Chem. Biodivers. 2023, 20, e202200911. [Google Scholar] [CrossRef]
  59. Skou, J. Chr. Further Investigations on a Mg++ + Na+-Activated Adenosintriphosphatase, Possibly Related to the Active, Linked Transport of Na+ and K+ across the Nerve Membrane. Biochim. Biophys. Acta 1960, 42, 6–23. [Google Scholar] [CrossRef]
  60. Blaustein, M.P.; Chen, L.; Hamlyn, J.M.; Leenen, F.H.H.; Lingrel, J.B.; Wier, W.G.; Zhang, J. Pivotal Role of A2 Na(+) Pumps and Their High Affinity Ouabain Binding Site in Cardiovascular Health and Disease. J. Physiol. 2016, 594, 6079–6103. [Google Scholar] [CrossRef]
  61. McDermott, J.P.; Numata, S.; Blanco, G. Na,K-ATPase Atp1a4 Isoform Is Important for Maintaining Sperm Flagellar Shape. J. Assist. Reprod. Genet. 2021, 38, 1493–1505. [Google Scholar] [CrossRef]
  62. Arystarkhova, E.; Sweadner, K.J. Na,K-ATPase Expression Can Be Limited Post-Transcriptionally: A Test of the Role of the Beta Subunit, and a Review of Evidence. Int. J. Mol. Sci. 2024, 25, 7414. [Google Scholar] [CrossRef]
  63. Geering, K. Functional Roles of Na,K-ATPase Subunits. Curr. Opin. Nephrol. Hypertens. 2008, 17, 526. [Google Scholar] [CrossRef]
  64. Kanai, R.; Cornelius, F.; Ogawa, H.; Motoyama, K.; Vilsen, B.; Toyoshima, C. Binding of Cardiotonic Steroids to Na+,K+-ATPase in the E2P State. Proc. Natl. Acad. Sci. USA 2021, 118, e2020438118. [Google Scholar] [CrossRef]
  65. 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]
  66. Tverskoi, A.M.; Poluektov, Y.M.; Klimanova, E.A.; Mitkevich, V.A.; Makarov, A.A.; Orlov, S.N.; Petrushanko, I.Y.; Lopina, O.D. Depth of the Steroid Core Location Determines the Mode of Na,K-ATPase Inhibition by Cardiotonic Steroids. Int. J. Mol. Sci. 2021, 22, 13268. [Google Scholar] [CrossRef]
  67. Pierre, S.V.; Xie, Z. The Na,K-ATPase Receptor Complex. Cell Biochem. Biophys. 2006, 46, 303–315. [Google Scholar] [CrossRef] [PubMed]
  68. Askari, A. The Other Functions of the Sodium Pump. Cell Calcium 2019, 84, 102105. [Google Scholar] [CrossRef] [PubMed]
  69. Pierre, S.V.; Blanco, G. Na/K-ATPase Ion Transport and Receptor-Mediated Signaling Pathways. J. Membr. Biol. 2021, 254, 443–446. [Google Scholar] [CrossRef]
  70. Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef]
  71. Sehnal, D.; Bittrich, S.; Deshpande, M.; Svobodová, R.; Berka, K.; Bazgier, V.; Velankar, S.; Burley, S.K.; Koča, J.; Rose, A.S. Mol* Viewer: Modern Web App for 3D Visualization and Analysis of Large Biomolecular Structures. Nucleic Acids Res. 2021, 49, W431–W437. [Google Scholar] [CrossRef]
  72. Kanai, R.; Cornelius, F.; Vilsen, B.; Toyoshima, C. Cryoelectron Microscopy of Na+,K+-ATPase in the Two E2P States with and without Cardiotonic Steroids. Proc. Natl. Acad. Sci. USA 2022, 119, e2123226119. [Google Scholar] [CrossRef]
  73. 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]
  74. Xie, Z. Molecular Mechanisms of Na/K-ATPase-Mediated Signal Transduction. Ann. N. Y. Acad. Sci. 2003, 986, 497–503. [Google Scholar] [CrossRef]
  75. Mohammadi, K.; Kometiani, P.; Xie, Z.; Askari, A. Role of Protein Kinase C in the Signal Pathways That Link Na+/K+-ATPase to ERK1/2. J. Biol. Chem. 2001, 276, 42050–42056. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, J.; Tian, J.; Haas, M.; Shapiro, J.I.; Askari, A.; Xie, Z. Ouabain Interaction with Cardiac Na+/K+-ATPase Initiates Signal Cascades Independent of Changes in Intracellular Na+ and Ca2+ Concentrations. J. Biol. Chem. 2000, 275, 27838–27844. [Google Scholar] [CrossRef] [PubMed]
  77. Aperia, A.; Brismar, H.; Uhlén, P. Mending Fences: Na,K-ATPase Signaling via Ca2+ in the Maintenance of Epithelium Integrity. Cell Calcium 2020, 88, 102210. [Google Scholar] [CrossRef] [PubMed]
  78. Wu, J.; Akkuratov, E.E.; Bai, Y.; Gaskill, C.M.; Askari, A.; Liu, L. Cell Signaling Associated with Na(+)/K(+)-ATPase: Activation of Phosphatidylinositide 3-Kinase IA/Akt by Ouabain Is Independent of Src. Biochemistry 2013, 52, 9059–9067. [Google Scholar] [CrossRef]
  79. Aizman, O.; Uhlén, P.; Lal, M.; Brismar, H.; Aperia, A. Ouabain, a Steroid Hormone That Signals with Slow Calcium Oscillations. Proc. Natl. Acad. Sci. USA 2001, 98, 13420–13424. [Google Scholar] [CrossRef]
  80. Zhang, S.; Malmersjö, S.; Li, J.; Ando, H.; Aizman, O.; Uhlén, P.; Mikoshiba, K.; Aperia, A. Distinct Role of the N-Terminal Tail of the Na,K-ATPase Catalytic Subunit as a Signal Transducer. J. Biol. Chem. 2006, 281, 21954–21962. [Google Scholar] [CrossRef]
  81. Panizza, E.; Zhang, L.; Fontana, J.M.; Hamada, K.; Svensson, D.; Akkuratov, E.E.; Scott, L.; Mikoshiba, K.; Brismar, H.; Lehtiö, J.; et al. Ouabain-Regulated Phosphoproteome Reveals Molecular Mechanisms for Na+, K+–ATPase Control of Cell Adhesion, Proliferation, and Survival. FASEB J. 2019, 33, 10193. [Google Scholar] [CrossRef]
  82. Pratt, R.D.; Brickman, C.R.; Cottrill, C.L.; Shapiro, J.I.; Liu, J. The Na/K-ATPase Signaling: From Specific Ligands to General Reactive Oxygen Species. Int. J. Mol. Sci. 2018, 19, 2600. [Google Scholar] [CrossRef]
  83. Huang, W.H.; Wang, Y.; Askari, A. (Na+ + K+)-ATPase: Inactivation and Degradation Induced by Oxygen Radicals. Int. J. Biochem. 1992, 24, 621–626. [Google Scholar] [CrossRef]
  84. Orlov, S.N.; Klimanova, E.A.; Tverskoi, A.M.; Vladychenskaya, E.A.; Smolyaninova, L.V.; Lopina, O.D. Na+i,K+i-Dependent and -Independent Signaling Triggered by Cardiotonic Steroids: Facts and Artifacts. Mol. J. Synth. Chem. Nat. Prod. Chem. 2017, 22, 635. [Google Scholar] [CrossRef]
  85. Fedorov, D.A.; Sidorenko, S.V.; Yusipovich, A.I.; Parshina, E.Y.; Tverskoi, A.M.; Abramicheva, P.A.; Maksimov, G.V.; Orlov, S.N.; Lopina, O.D.; Klimanova, E.A. Na+ i/K+ i Imbalance Contributes to Gene Expression in Endothelial Cells Exposed to Elevated NaCl. Heliyon 2021, 7, e08088. [Google Scholar] [CrossRef] [PubMed]
  86. Klimanova, E.A.; Tverskoi, A.M.; Koltsova, S.V.; Sidorenko, S.V.; Lopina, O.D.; Tremblay, J.; Hamet, P.; Kapilevich, L.V.; Orlov, S.N. Time- and Dose Dependent Actions of Cardiotonic Steroids on Transcriptome and Intracellular Content of Na+ and K+: A Comparative Analysis. Sci. Rep. 2017, 7, 45403. [Google Scholar] [CrossRef] [PubMed]
  87. Lopina, O.D.; Fedorov, D.A.; Sidorenko, S.V.; Bukach, O.V.; Klimanova, E.A. Sodium Ions as Regulators of Transcription in Mammalian Cells. Biochem. Mosc. 2022, 87, 789–799. [Google Scholar] [CrossRef] [PubMed]
  88. Cereijido, M.; Contreras, R.G.; Shoshani, L.; Larre, I. The Na+-K+-ATPase as Self-Adhesion Molecule and Hormone Receptor. Am. J. Physiol. Cell Physiol. 2012, 302, C473–C481. [Google Scholar] [CrossRef]
  89. Shoshani, L.; Contreras, R.G.; Roldán, M.L.; Moreno, J.; Lázaro, A.; Balda, M.S.; Matter, K.; Cereijido, M. The Polarized Expression of Na+,K+-ATPase in Epithelia Depends on the Association between Beta-Subunits Located in Neighboring Cells. Mol. Biol. Cell 2005, 16, 1071–1081. [Google Scholar] [CrossRef]
  90. Vagin, O.; Dada, L.A.; Tokhtaeva, E.; Sachs, G. The Na-K-ATPase A1β1 Heterodimer as a Cell Adhesion Molecule in Epithelia. Am. J. Physiol. Cell Physiol. 2012, 302, C1271–C1281. [Google Scholar] [CrossRef]
  91. Páez, O.; Martínez-Archundia, M.; Villegas-Sepúlveda, N.; Roldan, M.L.; Correa-Basurto, J.; Shoshani, L. A Model for the Homotypic Interaction between Na(+),K(+)-ATPase β(1) Subunits Reveals the Role of Extracellular Residues 221-229 in Its Ig-Like Domain. Int. J. Mol. Sci. 2019, 20, 4538. [Google Scholar] [CrossRef]
  92. Padilla-Benavides, T.; Roldán, M.L.; Larre, I.; Flores-Benitez, D.; Villegas-Sepúlveda, N.; Contreras, R.G.; Cereijido, M.; Shoshani, L. The Polarized Distribution of Na+,K+-ATPase: Role of the Interaction between {beta} Subunits. Mol. Biol. Cell 2010, 21, 2217–2225. [Google Scholar] [CrossRef]
  93. Tokhtaeva, E.; Sachs, G.; Souda, P.; Bassilian, S.; Whitelegge, J.P.; Shoshani, L.; Vagin, O. Epithelial Junctions Depend on Intercellular Trans-Interactions between the Na,K-ATPase Β1 Subunits. J. Biol. Chem. 2011, 286, 25801–25812. [Google Scholar] [CrossRef]
  94. Contreras, R.G.; Shoshani, L.; Flores-Maldonado, C.; Lázaro, A.; Cereijido, M. Relationship between Na(+),K(+)-ATPase and Cell Attachment. J. Cell Sci. 1999, 112 Pt 23, 4223–4232. [Google Scholar] [CrossRef]
  95. Lobato-Álvarez, J.A.; Roldán, M.L.; López-Murillo, T.D.C.; González-Ramírez, R.; Bonilla-Delgado, J.; Shoshani, L. The Apical Localization of Na(+), K(+)-ATPase in Cultured Human Retinal Pigment Epithelial Cells Depends on Expression of the β(2) Subunit. Front. Physiol. 2016, 7, 450. [Google Scholar] [CrossRef] [PubMed]
  96. Vilchis-Nestor, C.A.; Roldán, M.L.; Leonardi, A.; Navea, J.G.; Padilla-Benavides, T.; Shoshani, L. Ouabain Enhances Cell-Cell Adhesion Mediated by β(1) Subunits of the Na(+),K(+)-ATPase in CHO Fibroblasts. Int. J. Mol. Sci. 2019, 20, 2111. [Google Scholar] [CrossRef] [PubMed]
  97. Gloor, S.; Antonicek, H.; Sweadner, K.J.; Pagliusi, S.; Frank, R.; Moos, M.; Schachner, M. The Adhesion Molecule on Glia (AMOG) Is a Homologue of the Beta Subunit of the Na,K-ATPase. J. Cell Biol. 1990, 110, 165–174. [Google Scholar] [CrossRef] [PubMed]
  98. Müller-Husmann, G.; Gloor, S.; Schachner, M. Functional Characterization of Beta Isoforms of Murine Na,K-ATPase. Th e Adhesion Molecule on Glia (AMOG/Beta 2), but Not Beta 1, Promotes Ne Urite Outgrowth. J. Biol. Chem. 1993, 268, 26260–26267. [Google Scholar] [CrossRef]
  99. Roldán, M.L.; Ramírez-Salinas, G.L.; Martinez-Archundia, M.; Cuellar-Perez, F.; Vilchis-Nestor, C.A.; Cancino-Diaz, J.C.; Shoshani, L. The β(2)-Subunit (AMOG) of Human Na(+), K(+)-ATPase Is a Homophilic Adhesion Molecule. Int. J. Mol. Sci. 2022, 23, 7753. [Google Scholar] [CrossRef]
  100. Antonicek, H.; Persohn, E.; Schachner, M. Biochemical and Functional Characterization of a Novel Neuron-Glia Adhesion Molecule That Is Involved in Neuronal Migration. J. Cell Biol. 1987, 104, 1587–1595. [Google Scholar] [CrossRef]
  101. Hossan, M.S.; Chan, Z.-Y.; Collins, H.M.; Shipton, F.N.; Butler, M.S.; Rahmatullah, M.; Lee, J.B.; Gershkovich, P.; Kagan, L.; Khoo, T.-J.; et al. Cardiac Glycoside Cerberin Exerts Anticancer Activity through PI3K/AKT/mTOR Signal Transduction Inhibition. Cancer Lett. 2019, 453, 57–73. [Google Scholar] [CrossRef]
  102. Chen, G.; Zhang, H.; Sun, H.; Ding, X.; Liu, G.; Yang, F.; Feng, G.; Dong, X.; Zhu, Y.; Wang, X.; et al. Bufalin Targeting BFAR Inhibits the Occurrence and Metastasis of Gastric Cancer through PI3K/AKT/mTOR Signal Pathway. Apoptosis Int. J. Program. Cell Death 2023, 28, 1390–1405. [Google Scholar] [CrossRef]
  103. Reddy, D.; Ghosh, P.; Kumavath, R. Strophanthidin Attenuates MAPK, PI3K/AKT/mTOR, and Wnt/β-Catenin Signaling Pathways in Human Cancers. Front. Oncol. 2019, 9, 1469. [Google Scholar] [CrossRef]
  104. Xiang, C.; Cao, J.; Hu, R.; Li, K.; Meng, T.; Xia, Y.; Meng, Q.; Liu, K.; Liu, L.; Zhu, X. Oleandrin Inhibits Osteoclast Differentiation by Targeting the LRP4/MAPK/NF-κB Signalling Pathway to Treat Osteoporosis. Int. Immunopharmacol. 2025, 148, 114073. [Google Scholar] [CrossRef]
  105. Coleman, D.T.; Gray, A.L.; Stephens, C.A.; Scott, M.L.; Cardelli, J.A. Repurposed Drug Screen Identifies Cardiac Glycosides as Inhibitors of TGF-β-Induced Cancer-Associated Fibroblast Differentiation. Oncotarget 2016, 7, 32200–32209. [Google Scholar] [CrossRef] [PubMed]
  106. Yun, W.; Qian, L.; Yuan, R.; Xu, H. Periplocymarin Protects against Myocardial Fibrosis Induced by β-Adrenergic Activation in Mice. Biomed. Pharmacother. 2021, 139, 111562. [Google Scholar] [CrossRef] [PubMed]
  107. Cowman, S.J.; Koh, M.Y. Revisiting the HIF Switch in the Tumor and Its Immune Microenvironment. Trends Cancer 2022, 8, 28–42. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, H.; Qian, D.Z.; Tan, Y.S.; Lee, K.; Gao, P.; Ren, Y.R.; Rey, S.; Hammers, H.; Chang, D.; Pili, R.; et al. Digoxin and Other Cardiac Glycosides Inhibit HIF-1alpha Synthesis and Block Tumor Growth. Proc. Natl. Acad. Sci. USA 2008, 105, 19579–19586. [Google Scholar] [CrossRef]
  109. Lee, D.-H.; Cheul Oh, S.; Giles, A.J.; Jung, J.; Gilbert, M.R.; Park, D.M. Cardiac Glycosides Suppress the Maintenance of Stemness and Malignancy via Inhibiting HIF-1α in Human Glioma Stem Cells. Oncotarget 2017, 8, 40233–40245. [Google Scholar] [CrossRef]
  110. Yuan, Z.; Liu, C.; Sun, Y.; Li, Y.; Wu, H.; Ma, S.; Shang, J.; Zhan, Y.; Yin, P.; Gao, F. Bufalin Exacerbates Photodynamic Therapy of Colorectal Cancer by Targeting SRC-3/HIF-1α Pathway. Int. J. Pharm. 2022, 624, 122018. [Google Scholar] [CrossRef]
  111. 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]
  112. Zheng, Z.; Zhou, Z.; Zhang, Q.; Zhou, X.; Yang, J.; Yang, M.-R.; Zhu, G.-Y.; Jiang, Z.-H.; Li, T.; Lin, Q.; et al. Non-Classical Cardenolides from Calotropis Gigantea Exhibit Anticancer Effect as HIF-1 Inhibitors. Bioorganic Chem. 2021, 109, 104740. [Google Scholar] [CrossRef]
  113. Zhong, X.; Feng, W.; Liu, L.; Liu, Q.; Xu, Q.; Liu, M.; Liu, X.; Xu, S.; Deng, M.; Lin, C. Periplogenin Inhibits Pathologic Synovial Proliferation and Infiltration in Rheumatoid Arthritis by Regulating the JAK2/3-STAT3 Pathway. Int. Immunopharmacol. 2024, 128, 111487. [Google Scholar] [CrossRef]
  114. Fan, C.-L.; Liang, S.; Ye, M.-N.; Cai, W.-J.; Chen, M.; Hou, Y.-L.; Guo, J.; Dai, Y. Periplocymarin Alleviates Pathological Cardiac Hypertrophy via Inhibiting the JAK2/STAT3 Signalling Pathway. J. Cell. Mol. Med. 2022, 26, 2607–2619. [Google Scholar] [CrossRef]
  115. Kong, W.-S.; Shen, F.-X.; Xie, R.-F.; Zhou, G.; Feng, Y.-M.; Zhou, X. Bufothionine Induces Autophagy in H22 Hepatoma-Bearing Mice by Inhibiting JAK2/STAT3 Pathway, a Possible Anti-Cancer Mechanism of Cinobufacini. J. Ethnopharmacol. 2021, 270, 113848. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, Z.H.; Li, M.Y.; Wang, Z.; Zuo, H.X.; Wang, J.Y.; Xing, Y.; Jin, C.; Xu, G.; Piao, L.; Piao, H.; et al. Convallatoxin Promotes Apoptosis and Inhibits Proliferation and Angiogenesis through Crosstalk between JAK2/STAT3 (T705) and mTOR/STAT3 (S727) Signaling Pathways in Colorectal Cancer. Phytomedicine Int. J. Phytother. Phytopharm. 2020, 68, 153172. [Google Scholar] [CrossRef] [PubMed]
  117. Reddy, D.; Kumavath, R.; Tan, T.Z.; Ampasala, D.R.; Kumar, A.P. Peruvoside Targets Apoptosis and Autophagy through MAPK Wnt/β-Catenin and PI3K/AKT/mTOR Signaling Pathways in Human Cancers. Life Sci. 2020, 241, 117147. [Google Scholar] [CrossRef] [PubMed]
  118. Li, X.; Zheng, J.; Chen, S.; Meng, F.-D.; Ning, J.; Sun, S.-L. Oleandrin, a Cardiac Glycoside, Induces Immunogenic Cell Death via the PERK/elF2α/ATF4/CHOP Pathway in Breast Cancer. Cell Death Dis. 2021, 12, 314. [Google Scholar] [CrossRef]
  119. Zhao, W.; Li, G.; Zhang, Q.; Chen, M.; He, L.; Wu, Z.; Zhang, Y.; Fan, M.; Liang, Y.; Zhang, W.; et al. Cardiac Glycoside Neriifolin Exerts Anti-Cancer Activity in Prostate Cancer Cells by Attenuating DNA Damage Repair through Endoplasmic Reticulum Stress. Biochem. Pharmacol. 2023, 209, 115453. [Google Scholar] [CrossRef]
  120. Pavlovic, D. Endogenous Cardiotonic Steroids and Cardiovascular Disease, Where to Next? Cell Calcium 2020, 86, 102156. [Google Scholar] [CrossRef]
  121. Blaustein, M.P.; Hamlyn, J.M. Ouabain, Endogenous Ouabain and Ouabain-like Factors: The Na+ Pump/Ouabain Receptor, Its Linkage to NCX, and Its Myriad Functions. Cell Calcium 2020, 86, 102159. [Google Scholar] [CrossRef]
  122. Bejček, J.; Jurášek, M.; Spiwok, V.; Rimpelová, S. Quo Vadis Cardiac Glycoside Research? Toxins 2021, 13, 344. [Google Scholar] [CrossRef]
  123. Irgashev, I. Features and Benefits of Cardiac Glycosides in Chronic Heart Failure. Mod. Sci. Res. 2025, 4, 1292–1303. [Google Scholar]
  124. Riaz, T.; Akram, M.; Laila, U.; Zainab, R.; Khalil, M.T.; Iftikhar, M.; Ozdemir, F.A.; Sołowski, G.; Altable, M.; Sfera, A.; et al. Therapeutic Applications of Glycosides Obtained from Medicinal Plants. Int. Arch. Integr. Med. 2023, 10, 30–38. [Google Scholar]
  125. Khandelwal, R.; Vagha, J.D.; Meshram, R.J.; Patel, A. A Comprehensive Review on Unveiling the Journey of Digoxin: Past, Present, and Future Perspectives. Cureus 2024, 16, e56755. [Google Scholar] [CrossRef] [PubMed]
  126. Vishram-Nielsen, J.K.K.; Tomasoni, D.; Gustafsson, F.; Metra, M. Contemporary Drug Treatment of Advanced Heart Failure with Reduced Ejection Fraction. Drugs 2022, 82, 375–405. [Google Scholar] [CrossRef] [PubMed]
  127. Makhsumov, S.M.; Zayseva, O.A.; Djanaev, G.Y. Drugs Used in Heart Failure. J. Appl. Med. Sci. 2024, 7, 15–20. [Google Scholar]
  128. Miró, Ò.; Martín Mojarro, E.; Lopez-Ayala, P.; Llorens, P.; Gil, V.; Alquézar-Arbé, A.; Bibiano, C.; Pavón, J.; Massó, M.; Strebel, I.; et al. Association of Intravenous Digoxin Use in Acute Heart Failure with Rapid Atrial Fibrillation and Short-Term Mortality According to Patient Age, Renal Function, and Serum Potassium. Eur. J. Emerg. Med. Off. J. Eur. Soc. Emerg. Med. 2024, 31, 347–355. [Google Scholar] [CrossRef]
  129. Garg, U.; Christian, M. 5 Emergency and Intensive Care Management: For Acute Poisonings and Toxicities. In Interpreting Laboratory Tests in Intensive Care; CRC Press: Boca Raton, FL, USA, 2024; ISBN 978-1-003-44971-3. [Google Scholar]
  130. Khalaf, F.K.; Dube, P.; Mohamed, A.; Tian, J.; Malhotra, D.; Haller, S.T.; Kennedy, D.J. Cardiotonic Steroids and the Sodium Trade Balance: New Insights into Trade-Off Mechanisms Mediated by the Na+/K+-ATPase. Int. J. Mol. Sci. 2018, 19, 2576. [Google Scholar] [CrossRef]
  131. Manunta, P.; Hamilton, J.; Rogowski, A.C.; Hamilton, B.P.; Hamlyn, J.M. Chronic Hypertension Induced by Ouabain but Not Digoxin in the Rat: Antihypertensive Effect of Digoxin and Digitoxin. Hypertens. Res. 2000, 23, S77–S85. [Google Scholar] [CrossRef]
  132. Blaustein, M.P.; Gottlieb, S.S.; Hamlyn, J.M.; Leenen, F.H.H. Whither Digitalis? What We Can Still Learn from Cardiotonic Steroids about Heart Failure and Hypertension. Am. J. Physiol. Heart Circ. Physiol. 2022, 323, H1281–H1295. [Google Scholar] [CrossRef]
  133. Manunta, P.; Hamilton, B.P.; Hamlyn, J.M. Salt Intake and Depletion Increase Circulating Levels of Endogenous Ouabain in Normal Men. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2006, 290, R553–R559. [Google Scholar] [CrossRef]
  134. Manfrini, V.; Badagliacca, R.; Messaggio, E.; Poscia, R.; Torre, R.; Manunta, P.; Vizza, C.D. Exploratory Study on the Endogenous Ouabain in Idiopathic Pulmonary Arterial Hypertension Patients. Ann. Ist. Super. Sanita 2023, 59, 76–79. [Google Scholar] [CrossRef]
  135. Tomaschitz, A.; Piecha, G.; Ritz, E.; Meinitzer, A.; Haas, J.; Pieske, B.; Wiecek, A.; Rus-Machan, J.; Toplak, H.; März, W.; et al. Marinobufagenin in Essential Hypertension and Primary Aldosteronism: A Cardiotonic Steroid with Clinical and Diagnostic Implications. Clin. Exp. Hypertens. 2015, 37, 108–115. [Google Scholar] [CrossRef]
  136. Tian, J.; Haller, S.; Periyasamy, S.; Brewster, P.; Zhang, H.; Adlakha, S.; Fedorova, O.V.; Xie, Z.-J.; Bagrov, A.Y.; Shapiro, J.I.; et al. Renal Ischemia Regulates Marinobufagenin Release in Humans. Hypertension 2010, 56, 914–919. [Google Scholar] [CrossRef] [PubMed]
  137. Fedorova, O.V.; Shapiro, J.I.; Bagrov, A.Y. Endogenous Cardiotonic Steroids and Salt-Sensitive Hypertension. Biochim. Biophys. Acta 2010, 1802, 1230–1236. [Google Scholar] [CrossRef] [PubMed]
  138. Aung, K.; Ream-Winnick, S.; Lane, M.; Akinlusi, I.; Shi, T.; Htay, T. Sodium Homeostasis and Hypertension. Curr. Cardiol. Rep. 2023, 25, 1123–1129. [Google Scholar] [CrossRef] [PubMed]
  139. Łabno-Kirszniok, K.; Kujawa-Szewieczek, A.; Wiecek, A.; Piecha, G. The Effects of Short-Term Changes in Sodium Intake on Plasma Marinobufagenin Levels in Patients with Primary Salt-Sensitive and Salt-Insensitive Hypertension. Nutrients 2021, 13, 1502. [Google Scholar] [CrossRef]
  140. Schoner, W.; Scheiner-Bobis, G. Role of Endogenous Cardiotonic Steroids in Sodium Homeostasis. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. Eur. Ren. Assoc. 2008, 23, 2723–2729. [Google Scholar] [CrossRef]
  141. Cereijido, M.; Contreras, R.G.; Shoshani, L. Cell Adhesion, Polarity, and Epithelia in the Dawn of Metazoans. Physiol. Rev. 2004, 84, 1229–1262. [Google Scholar] [CrossRef]
  142. Cereijido, M.; Contreras, R.G.; Shoshani, L.; Flores-Benitez, D.; Larre, I. Tight Junction and Polarity Interaction in the Transporting Epithelial Phenotype. Biochim. Biophys. Acta 2008, 1778, 770–793. [Google Scholar] [CrossRef]
  143. Rincon-Heredia, R.; Flores-Benitez, D.; Flores-Maldonado, C.; Bonilla-Delgado, J.; García-Hernández, V.; Verdejo-Torres, O.; Castillo, A.M.; Larré, I.; Poot-Hernández, A.C.; Franco, M.; et al. Ouabain Induces Endocytosis and Degradation of Tight Junction Proteins through ERK1/2-Dependent Pathways. Exp. Cell Res. 2014, 320, 108–118. [Google Scholar] [CrossRef]
  144. Contreras, R.G.; Flores-Beni Tez, D.; Flores-Maldonado, C.; Larre, I.; Shoshani, L.; Cereijido, M. Na+,K+-ATPase and Hormone Ouabain:New Roles for an Old Enzyme and an Old Inhibitor. Cell. Mol. Biol. Noisy-Gd. Fr. 2006, 52, 31–40. [Google Scholar]
  145. Larre, I.; Lazaro, A.; Contreras, R.G.; Balda, M.S.; Matter, K.; Flores-Maldonado, C.; Ponce, A.; Flores-Benitez, D.; Rincon-Heredia, R.; Padilla-Benavides, T.; et al. Ouabain Modulates Epithelial Cell Tight Junction. Proc. Natl. Acad. Sci. USA 2010, 107, 11387–11392. [Google Scholar] [CrossRef]
  146. Castillo, A.; Ortuño-Pineda, C.; Flores-Maldonado, C.; Larre, I.; Martínez Rendón, J.; Hinojosa, L.; Ponce, A.; Ogazón, A.; Serrano, M.; Valdes, J.; et al. Ouabain Modulates the Adherens Junction in Renal Epithelial Cells. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2019, 52, 1381–1397. [Google Scholar] [CrossRef]
  147. Ponce, A.; Larre, I.; Castillo, A.; García-Villegas, R.; Romero, A.; Flores-Maldonado, C.; Martinez-Rendón, J.; Contreras, R.G.; Cereijido, M. Ouabain Increases Gap Junctional Communication in Epithelial Cells. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2014, 34, 2081–2090. [Google Scholar] [CrossRef] [PubMed]
  148. Ponce, A.; Larre, I.; Castillo, A.; Flores-Maldonado, C.; Verdejo-Torres, O.; Contreras, R.G.; Cereijido, M. Ouabain Modulates the Distribution of Connexin 43 in Epithelial Cells. Cell. Physiol. Biochem. 2016, 39, 1329–1338. [Google Scholar] [CrossRef] [PubMed]
  149. Serrano-Rubi, M.; Jimenez, L.; Martinez-Rendon, J.; Cereijido, M.; Ponce, A. Ouabain Promotes Gap Junctional Intercellular Communication in Cancer Cells. Int. J. Mol. Sci. 2020, 22, 358. [Google Scholar] [CrossRef]
  150. Ogazon Del Toro, A.; Jimenez, L.; Hinojosa, L.; Martínez-Rendón, J.; Castillo, A.; Cereijido, M.; Ponce, A. Influence of Endogenous Cardiac Glycosides, Digoxin, and Marinobufagenin in the Physiology of Epithelial Cells. Cardiol. Res. Pract. 2019, 2019, 8646787. [Google Scholar] [CrossRef]
  151. Larre, I.; Castillo, A.; Flores-Maldonado, C.; Contreras, R.G.; Galvan, I.; Muñoz-Estrada, J.; Cereijido, M. Ouabain Modulates Ciliogenesis in Epithelial Cells. Proc. Natl. Acad. Sci. USA 2011, 108, 20591–20596. [Google Scholar] [CrossRef]
  152. Cereijido, M.; Jimenez, L.; Hinojosa, L.; Castillo, A.; Martínez-Rendon, J.; Ponce, A. Ouabain-Induced Changes in the Expression of Voltage-Gated Potassium Channels in Epithelial Cells Depend on Cell-Cell Contacts. Int. J. Mol. Sci. 2022, 23, 3257. [Google Scholar] [CrossRef]
  153. Ponce, A.; Larre, I.; Jimenez, L.; Roldán, M.L.; Shoshani, L.; Cereijido, M. Ouabain’s Influence on TRPV4 Channels of Epithelial Cells: An Exploration of TRPV4 Activity, Expression, and Signaling Pathways. Int. J. Mol. Sci. 2023, 24, 16687. [Google Scholar] [CrossRef]
  154. Ogazon Del Toro, A.; Jimenez, L.; Serrano Rubi, M.; Cereijido, M.; Ponce, A. Ouabain Enhances Gap Junctional Intercellular Communication by Inducing Paracrine Secretion of Prostaglandin E2. Int. J. Mol. Sci. 2021, 22, 6244. [Google Scholar] [CrossRef]
  155. Martínez-Rendón, J.; Hinojosa, L.; Xoconostle-Cázares, B.; Ramírez-Pool, J.A.; Castillo, A.; Cereijido, M.; Ponce, A. Ouabain Induces Transcript Changes and Activation of RhoA/ROCK Signaling in Cultured Epithelial Cells (MDCK). Curr. Issues Mol. Biol. 2023, 45, 7538–7556. [Google Scholar] [CrossRef]
  156. Dizon, D.S.; Kamal, A.H. Cancer Statistics 2024: All Hands on Deck. CA. Cancer J. Clin. 2024, 74, 8–9. [Google Scholar] [CrossRef] [PubMed]
  157. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [PubMed]
  158. Vishnoi, K.; Viswakarma, N.; Rana, A.; Rana, B. Transcription Factors in Cancer Development and Therapy. Cancers 2020, 12, 2296. [Google Scholar] [CrossRef] [PubMed]
  159. Stenkvist, B.; Bengtsson, E.; Eriksson, O.; Holmquist, J.; Nordin, B.; Westman-Naeser, S.; Eklund, G. Cardiac Glycosides and Breast Cancer. Lancet 1979, 313, 563. [Google Scholar] [CrossRef]
  160. Kumavath, R.; Paul, S.; Pavithran, H.; Paul, M.K.; Ghosh, P.; Barh, D.; Azevedo, V. Emergence of Cardiac Glycosides as Potential Drugs: Current and Future Scope for Cancer Therapeutics. Biomolecules 2021, 11, 1275. [Google Scholar] [CrossRef]
  161. Reddy, D.; Kumavath, R.; Barh, D.; Azevedo, V.; Ghosh, P. Anticancer and Antiviral Properties of Cardiac Glycosides: A Review to Explore the Mechanism of Actions. Molecules 2020, 25, 3596. [Google Scholar] [CrossRef]
  162. Ho, H.-Y.; Chen, M.-K.; Lin, C.-C.; Lo, Y.-S.; Chuang, Y.-C.; Hsieh, M.-J. Arenobufagin Induces Cell Apoptosis by Modulating the Cell Cycle Regulator Claspin and the JNK Pathway in Nasopharyngeal Carcinoma Cells. Expert Opin. Ther. Targets 2024, 28, 461–471. [Google Scholar] [CrossRef]
  163. Hu, R.; Tang, S.; Xiang, Y.; Qin, S. Arenobufagin Induces Ferroptosis in Glioblastoma Cells via Modulating the MiR-149-5p/AEBP1 Axis. J. Appl. Toxicol. JAT 2025, 45, 606–619. [Google Scholar] [CrossRef]
  164. Chen, L.; Mai, W.; Chen, M.; Hu, J.; Zhuo, Z.; Lei, X.; Deng, L.; Liu, J.; Yao, N.; Huang, M.; et al. Arenobufagin Inhibits Prostate Cancer Epithelial-Mesenchymal Transition and Metastasis by down-Regulating β-Catenin. Pharmacol. Res. 2017, 123, 130–142. [Google Scholar] [CrossRef]
  165. Yang, Y.; Liu, C.; Wang, M.; Cheng, H.; Wu, H.; Luo, S.; Zhang, M.; Duan, X.; Li, Q. Arenobufagin Regulates the P62-Keap1-Nrf2 Pathway to Induce Autophagy-Dependent Ferroptosis in HepG2 Cells. Naunyn. Schmiedebergs Arch. Pharmacol. 2024, 397, 4895–4909. [Google Scholar] [CrossRef]
  166. 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]
  167. Qi, Y.; Wu, H.; Zhu, T.; Liu, Z.; Liu, C.; Yan, C.; Wu, Z.; Xu, Y.; Bai, Y.; Yang, L.; et al. Acetyl-Cinobufagin Suppresses Triple-Negative Breast Cancer Progression by Inhibiting the STAT3 Pathway. Aging 2023, 15, 8258–8274. [Google Scholar] [CrossRef] [PubMed]
  168. Chen, J.; Wu, C.; Yu, K.; Liu, J.; Yang, J.; Li, W.; Tang, X.; Shi, Y.; Xu, K.; Chen, Y.; et al. Bufalin Targets the SRC-3/c-Myc Pathway in Chemoresistant Cells to Regulate Metastasis Induced by Chemoresistance in Colorectal Cancer. J. Cancer Res. Clin. Oncol. 2025, 151, 71. [Google Scholar] [CrossRef] [PubMed]
  169. Shang, J.; Xia, Q.; Sun, Y.; Wang, H.; Chen, J.; Li, Y.; Gao, F.; Yin, P.; Yuan, Z. Bufalin-Loaded Multifunctional Photothermal Nanoparticles Inhibit the Anaerobic Glycolysis by Targeting SRC-3/HIF-1α Pathway for Improved Mild Photothermal Therapy in CRC. Int. J. Nanomed. 2024, 19, 7831–7850. [Google Scholar] [CrossRef]
  170. Zheng, Y.-D.; He, Z.; Su, Z.-C.; Wang, H.; Jiang, X.-H.; Fang, X.; Lu, S.-L.; Li, Y. Bufalin Induces Apoptosis and Autophagy via the Ca2+/CaMKKβ/AMPK/Beclin1 Signaling Pathway in Osteosarcoma Cells. Cell Biol. Int. 2023, 47, 1344–1353. [Google Scholar] [CrossRef]
  171. Zhang, H.; Dong, X.; Ding, X.; Liu, G.; Yang, F.; Song, Q.; Sun, H.; Chen, G.; Li, S.; Li, Y.; et al. Bufalin Targeting CAMKK2 Inhibits the Occurrence and Development of Intrahepatic Cholangiocarcinoma through Wnt/β-Catenin Signal Pathway. J. Transl. Med. 2023, 21, 900. [Google Scholar] [CrossRef]
  172. Qian, Z.; Tian, X.; Miao, Y.; Xu, X.; Cheng, X.; Wu, M.; Yu, Y. Bufalin Inhibits the Proliferation of Lung Cancer Cells by Suppressing Hippo-YAP Pathway. Cell. Signal. 2023, 109, 110746. [Google Scholar] [CrossRef]
  173. Ju, Q.; Shi, Q.; Liu, C.; Fu, G.; Shi, H. Bufalin Suppresses Esophageal Squamous Cell Carcinoma Progression by Activating the PIAS3/STAT3 Signaling Pathway. J. Thorac. Dis. 2023, 15, 2141–2160. [Google Scholar] [CrossRef]
  174. Farooqi, A.A.; Rakhmetova, V.S.; Kapanova, G.; Tashenova, G.; Tulebayeva, A.; Akhenbekova, A.; Ibekenov, O.; Turgambayeva, A.; Xu, B. Bufalin-Mediated Regulation of Cell Signaling Pathways in Different Cancers: Spotlight on JAK/STAT, Wnt/β-Catenin, mTOR, TRAIL/TRAIL-R, and Non-Coding RNAs. Molecules 2023, 28, 2231. [Google Scholar] [CrossRef]
  175. Zhu, Z.; Liang, S.; Hong, Y.; Qi, Y.; Sun, Q.; Zhu, X.; Wei, Y.; Xu, Y.; Chen, Q. Bufotalin Enhances Apoptosis and TMZ Chemosensitivity of Glioblastoma Cells by Promoting Mitochondrial Dysfunction via AKT Signaling Pathway. Aging 2024, 16, 9264–9279. [Google Scholar] [CrossRef]
  176. Zhang, G.; Zhang, K.; Li, X.; Wang, X.; Li, G.; Wang, Y. Cinobufagin Enhances the Sensitivity of Cisplatin-Resistant Lung Cancer Cells to Chemotherapy by Inhibiting the PI3K/AKT and MAPK/ERK Pathways. J. Cell. Mol. Med. 2025, 29, e70501. [Google Scholar] [CrossRef] [PubMed]
  177. Hu, Y.; Luo, M. Cinobufotalin Regulates the USP36/c-Myc Axis to Suppress Malignant Phenotypes of Colon Cancer Cells in Vitro and in Vivo. Aging 2024, 16, 5526–5544. [Google Scholar] [CrossRef] [PubMed]
  178. Liu, X.; Geng, Z.; Ding, X.; Lou, Y.; Zhang, X. Convallatoxin Suppresses Osteosarcoma Cell Proliferation, Migration, Invasion, and Enhances Osteogenic Differentiation by Downregulating Parathyroid Hormone Receptor 1 (PTHR1) Expression and Inactivating Wnt/β-Catenin Pathway. Bioengineered 2022, 13, 13280–13292. [Google Scholar] [CrossRef] [PubMed]
  179. Zhan, Y.; Wang, R.; Huang, C.; Xu, X.; Xiao, X.; Wu, L.; Wei, J.; Long, T.; Gao, C. Digitoxin Inhibits ICC Cell Properties via the NF-κB/ST6GAL1 Signaling Pathway. Oncol. Rep. 2024, 52, 103. [Google Scholar] [CrossRef]
  180. Yun, H.H.; Kim, S.; Kuh, H.-J.; Lee, J.-H. Downregulation of BIS Sensitizes A549 Cells for Digoxin-Mediated Inhibition of Invasion and Migration by the STAT3-Dependent Pathway. Biochem. Biophys. Res. Commun. 2020, 524, 643–648. [Google Scholar] [CrossRef]
  181. Ma, K.; Zhang, C.; Li, W. Gamabufotalin Suppressed Osteosarcoma Stem Cells through the TGF-β/Periostin/PI3K/AKT Pathway. Chem. Biol. Interact. 2020, 331, 109275. [Google Scholar] [CrossRef]
  182. Lan, Y.-L.; Chen, C.; Wang, X.; Lou, J.-C.; Xing, J.-S.; Zou, S.; Hu, J.-L.; Lyu, W.; Zhang, B. Gamabufotalin Induces a Negative Feedback Loop Connecting ATP1A3 Expression and the AQP4 Pathway to Promote Temozolomide Sensitivity in Glioblastoma Cells by Targeting the Amino Acid Thr794. Cell Prolif. 2020, 53, e12732. [Google Scholar] [CrossRef]
  183. Reddy, D.; Kumavath, R.; Ghosh, P.; Barh, D. Lanatoside C Induces G2/M Cell Cycle Arrest and Suppresses Cancer Cell Growth by Attenuating MAPK, Wnt, JAK-STAT, and PI3K/AKT/mTOR Signaling Pathways. Biomolecules 2019, 9, 792. [Google Scholar] [CrossRef]
  184. Huang, S.; Huang, D.; Jin, Y.; Shao, C.; Su, X.; Yang, R.; Jiang, J.; Wu, J. Lanatoside C Inhibits Proliferation and Induces Apoptosis in Human Prostate Cancer Cells Through the TNF/IL-17 Signaling Pathway. Int. J. Mol. Sci. 2025, 26, 2558. [Google Scholar] [CrossRef]
  185. Hu, Q.-Y.; Zhang, X.-K.; Wang, J.-N.; Chen, H.-X.; He, L.-P.; Tang, J.-S.; Yao, X.-S.; Liu, J. Malayoside, a Cardenolide Glycoside Extracted from Antiaris Toxicaria Lesch, Induces Apoptosis in Human Non-Small Lung Cancer Cells via MAPK-Nur77 Signaling Pathway. Biochem. Pharmacol. 2021, 190, 114622. [Google Scholar] [CrossRef]
  186. Hu, X.; Chen, T.; Zhang, S.; Zhang, Q.; Li, C.; Wang, X. Antitumour Effect of Odoroside A and Its Derivative on Human Leukaemia Cells through the ROS/JNK Pathway. Basic Clin. Pharmacol. Toxicol. 2022, 130, 56–69. [Google Scholar] [CrossRef] [PubMed]
  187. Ko, Y.S.; Rugira, T.; Jin, H.; Park, S.W.; Kim, H.J. Oleandrin and Its Derivative Odoroside A, Both Cardiac Glycosides, Exhibit Anticancer Effects by Inhibiting Invasion via Suppressing the STAT-3 Signaling Pathway. Int. J. Mol. Sci. 2018, 19, 3350. [Google Scholar] [CrossRef] [PubMed]
  188. Zhai, J.; Dong, X.; Yan, F.; Guo, H.; Yang, J. Oleandrin: A Systematic Review of Its Natural Sources, Structural Properties, Detection Methods, Pharmacokinetics and Toxicology. Front. Pharmacol. 2022, 13, 822726. [Google Scholar] [CrossRef] [PubMed]
  189. Shen, J.-J.; Zhan, Y.-C.; Li, H.-Y.; Wang, Z. Ouabain Impairs Cancer Metabolism and Activates AMPK-Src Signaling Pathway in Human Cancer Cell Lines. Acta Pharmacol. Sin. 2020, 41, 110–118. [Google Scholar] [CrossRef]
  190. Lai, Y.; Chang, H.; Chen, H.; Chang, G.; Chen, J.J. Peruvoside Is a Novel Src Inhibitor That Suppresses NSCLC Cell Growth and Motility by Downregulating Multiple Src-EGFR-Related Pathways. Am. J. Cancer Res. 2022, 12, 2576–2593. [Google Scholar]
  191. Hu, Y.; Liu, F.; Jia, X.; Wang, P.; Gu, T.; Liu, H.; Liu, T.; Wei, H.; Chen, H.; Zhao, J.; et al. Periplogenin Suppresses the Growth of Esophageal Squamous Cell Carcinoma in Vitro and in Vivo by Targeting STAT3. Oncogene 2021, 40, 3942–3958. [Google Scholar] [CrossRef]
  192. Zhou, G.; Zhu, Z.; Li, L.; Ding, J. Resibufogenin Inhibits Ovarian Clear Cell Carcinoma (OCCC) Growth in Vivo, and Migration of OCCC Cells in Vitro, by down-Regulating the PI3K/AKT and Actin Cytoskeleton Signaling Pathways. Am. J. Transl. Res. 2019, 11, 6290–6303. [Google Scholar]
  193. Zhou, Y.; Hong, Z.; Jin, K.; Lin, C.; Xiang, J.; Ge, H.; Zheng, Z.; Shen, J.; Deng, S. Resibufogenin Inhibits the Malignant Characteristics of Multiple Myeloma Cells by Blocking the PI3K/Akt Signaling Pathway. Exp. Ther. Med. 2022, 24, 441. [Google Scholar] [CrossRef]
  194. Yang, T.; Jiang, Y.-X.; Wu, Y.; Lu, D.; Huang, R.; Wang, L.-L.; Wang, S.-Q.; Guan, Y.-Y.; Zhang, H.; Luan, X. Resibufogenin Suppresses Triple-Negative Breast Cancer Angiogenesis by Blocking VEGFR2-Mediated Signaling Pathway. Front. Pharmacol. 2021, 12, 682735. [Google Scholar] [CrossRef]
  195. Huang, J.; Deng, C.; Guo, T.; Chen, X.; Chen, P.; Du, S.; Lu, M. Cinobufotalin Induces Ferroptosis to Suppress Lung Cancer Cell Growth by lncRNA LINC00597/Hsa-miR-367-3p/TFRC Pathway via Resibufogenin. Anticancer Agents Med. Chem. 2023, 23, 717–725. [Google Scholar] [CrossRef]
  196. Qiang, L.-Z.; Fang, S.-Z. Telocinobufagin Suppresses Malignant Metastasis of Undifferentiated Thyroid Carcinoma via Modulation of the LARP1-mTOR Pathway. Kaohsiung J. Med. Sci. 2025, 41, e12934. [Google Scholar] [CrossRef] [PubMed]
  197. Shen, Y.; Cai, H.; Ma, S.; Zhu, W.; Zhao, H.; Li, J.; Ye, H.; Yang, L.; Zhao, C.; Huang, X.; et al. Telocinobufagin Has Antitumor Effects in Non-Small-Cell Lung Cancer by Inhibiting STAT3 Signaling. J. Nat. Prod. 2022, 85, 765–775. [Google Scholar] [CrossRef] [PubMed]
  198. Weng, J.-R.; Lin, W.-Y.; Bai, L.-Y.; Hu, J.-L.; Feng, C.-H. Antitumor Activity of the Cardiac Glycoside αlDiginoside by Modulating Mcl-1 in Human Oral Squamous Cell Carcinoma Cells. Int. J. Mol. Sci. 2020, 21, 7947. [Google Scholar] [CrossRef] [PubMed]
  199. Varela-Eirín, M.; Demaria, M. Cellular Senescence. Curr. Biol. 2022, 32, R448–R452. [Google Scholar] [CrossRef]
  200. Zhang, X.; Gao, Y.; Zhang, S.; Wang, Y.; Du, Y.; Hao, S.; Ni, T. The Regulation of Cellular Senescence in Cancer. Biomolecules 2025, 15, 448. [Google Scholar] [CrossRef]
  201. Martin, N.; Soriani, O.; Bernard, D. Cardiac Glycosides as Senolytic Compounds. Trends Mol. Med. 2020, 26, 243–245. [Google Scholar] [CrossRef]
  202. Triana-Martínez, F.; Picallos-Rabina, P.; Da Silva-Álvarez, S.; Pietrocola, F.; Llanos, S.; Rodilla, V.; Soprano, E.; Pedrosa, P.; Ferreirós, A.; Barradas, M.; et al. Identification and Characterization of Cardiac Glycosides as Senolytic Compounds. Nat. Commun. 2019, 10, 4731. [Google Scholar] [CrossRef]
  203. Guerrero, A.; Herranz, N.; Sun, B.; Wagner, V.; Gallage, S.; Guiho, R.; Wolter, K.; Pombo, J.; Irvine, E.E.; Innes, A.J.; et al. Cardiac Glycosides Are Broad-Spectrum Senolytics. Nat. Metab. 2019, 1, 1074–1088. [Google Scholar] [CrossRef]
  204. Smer-Barreto, V.; Quintanilla, A.; Elliott, R.J.R.; Dawson, J.C.; Sun, J.; Campa, V.M.; Lorente-Macías, Á.; Unciti-Broceta, A.; Carragher, N.O.; Acosta, J.C.; et al. Discovery of Senolytics Using Machine Learning. Nat. Commun. 2023, 14, 3445. [Google Scholar] [CrossRef]
  205. Takaya, K.; Asou, T.; Kishi, K. Identification of Resibufogenin, a Component of Toad Venom, as a Novel Senolytic Compound in Vitro and for Potential Skin Rejuvenation in Male Mice. Biogerontology 2023, 24, 889–900. [Google Scholar] [CrossRef]
  206. Ketelut-Carneiro, N.; Fitzgerald, K.A. Apoptosis, Pyroptosis, and Necroptosis—Oh My! The Many Ways a Cell Can Die. J. Mol. Biol. 2022, 434, 167378. [Google Scholar] [CrossRef] [PubMed]
  207. Mustafa, M.; Ahmad, R.; Tantry, I.Q.; Ahmad, W.; Siddiqui, S.; Alam, M.; Abbas, K.; Moinuddin; Hassan, M.I.; Habib, S.; et al. Apoptosis: A Comprehensive Overview of Signaling Pathways, Morphological Changes, and Physiological Significance and Therapeutic Implications. Cells 2024, 13, 1838. [Google Scholar] [CrossRef] [PubMed]
  208. Karati, D.; Kumar, D. Molecular Insight into the Apoptotic Mechanism of Cancer Cells: An Explicative Review. Curr. Mol. Pharmacol. 2024, 17, e18761429273223. [Google Scholar] [CrossRef] [PubMed]
  209. Ainembabazi, D.; Zhang, Y.; Turchi, J.J. The Mechanistic Role of Cardiac Glycosides in DNA Damage Response and Repair Signaling. Cell. Mol. Life Sci. CMLS 2023, 80, 250. [Google Scholar] [CrossRef]
  210. Škubník, J.; Svobodová Pavlíčková, V.; Psotová, J.; Rimpelová, S. Cardiac Glycosides as Autophagy Modulators. Cells 2021, 10, 3341. [Google Scholar] [CrossRef]
  211. Dong, Q.; Turdu, G.; Akber Aisa, H.; Yili, A. Arenobufagin, Isolated from Bufo Viridis Toad Venom, Inhibits A549 Cells Proliferation by Inducing Apoptosis and G2/M Cell Cycle Arrest. Toxicon Off. J. Int. Soc. Toxinology 2024, 240, 107641. [Google Scholar] [CrossRef]
  212. Wei, X.; Yang, J.; Mao, Y.; Zhao, H.; Si, N.; Wang, H.; Bian, B. Arenobufagin Inhibits the Phosphatidylinositol 3-Kinase/Protein Kinase B/Mammalian Target of Rapamycin Pathway and Induces Apoptosis and Autophagy in Pancreatic Cancer Cells. Pancreas 2020, 49, 261–272. [Google Scholar] [CrossRef]
  213. Li, Y.; Zhang, Y.; Wang, X.; Yang, Q.; Zhou, X.; Wu, J.; Yang, X.; Zhao, Y.; Lin, R.; Xie, Y.; et al. Bufalin Induces Mitochondrial Dysfunction and Promotes Apoptosis of Glioma Cells by Regulating Annexin A2 and DRP1 Protein Expression. Cancer Cell Int. 2021, 21, 424. [Google Scholar] [CrossRef]
  214. Pan, L.; Nie, L.; Yao, S.; Bi, A.; Ye, Y.; Wu, Y.; Tan, Z.; Wu, Z. Bufalin Exerts Antitumor Effects in Neuroblastoma via the Induction of Reactive Oxygen Species-mediated Apoptosis by Targeting the Electron Transport Chain. Int. J. Mol. Med. 2020, 46, 2137–2149. [Google Scholar] [CrossRef]
  215. LingHu, H.R.; Luo, H.; Gang, L. Bufalin Induces Glioma Cell Death by Apoptosis or Necroptosis. OncoTargets Ther. 2020, 13, 4767–4778. [Google Scholar] [CrossRef]
  216. Han, Q.; Zhang, C.; Zhang, Y.; Li, Y.; Wu, L.; Sun, X. Bufarenogin Induces Intrinsic Apoptosis via Bax and ANT Cooperation. Pharmacol. Res. Perspect. 2021, 9, e00694. [Google Scholar] [CrossRef] [PubMed]
  217. Park, S.J.; Jung, H.J. Bufotalin Suppresses Proliferation and Metastasis of Triple-Negative Breast Cancer Cells by Promoting Apoptosis and Inhibiting the STAT3/EMT Axis. Molecules 2023, 28, 6783. [Google Scholar] [CrossRef] [PubMed]
  218. Zhang, L.; Liang, B.; Xu, H.; Gong, Y.; Hu, W.; Jin, Z.; Wu, X.; Chen, X.; Li, M.; Shi, L.; et al. Cinobufagin Induces FOXO1-Regulated Apoptosis, Proliferation, Migration, and Invasion by Inhibiting G9a in Non-Small-Cell Lung Cancer A549 Cells. J. Ethnopharmacol. 2022, 291, 115095. [Google Scholar] [CrossRef] [PubMed]
  219. Bian, Y.; Xue, M.; Guo, X.; Jiang, W.; Zhao, Y.; Zhang, Z.; Wang, X.; Hu, Y.; Zhang, Q.; Dun, W.; et al. Cinobufagin Induces Acute Promyelocytic Leukaemia Cell Apoptosis and PML-RARA Degradation in a Caspase-Dependent Manner by Inhibiting the β-Catenin Signalling Pathway. Pharm. Biol. 2022, 60, 1801–1811. [Google Scholar] [CrossRef]
  220. Niu, J.; Wang, J.; Zhang, Q.; Zou, Z.; Ding, Y. Cinobufagin-Induced DNA Damage Response Activates G(2)/M Checkpoint and Apoptosis to Cause Selective Cytotoxicity in Cancer Cells. Cancer Cell Int. 2021, 21, 446. [Google Scholar] [CrossRef]
  221. Pan, Z.; Luo, Y.; Xia, Y.; Zhang, X.; Qin, Y.; Liu, W.; Li, M.; Liu, X.; Zheng, Q.; Li, D. Cinobufagin Induces Cell Cycle Arrest at the S Phase and Promotes Apoptosis in Nasopharyngeal Carcinoma Cells. Biomed. Pharmacother. 2020, 122, 109763. [Google Scholar] [CrossRef]
  222. Mi, C.; Cao, X.; Ma, K.; Wei, M.; Xu, W.; Lin, Y.; Zhang, J.; Wang, T.-Y. Digitoxin Promotes Apoptosis and Inhibits Proliferation and Migration by Reducing HIF-1α and STAT3 in KRAS Mutant Human Colon Cancer Cells. Chem. Biol. Interact. 2022, 351, 109729. [Google Scholar] [CrossRef]
  223. Hsieh, M.-J.; Lin, C.-C.; Lo, Y.-S.; Ho, H.-Y.; Chuang, Y.-C.; Chen, M.-K. Hellebrigenin Induces Oral Cancer Cell Apoptosis by Modulating MAPK Signalling and XIAP Expression. J. Cell. Mol. Med. 2024, 28, e18071. [Google Scholar] [CrossRef]
  224. Zhang, C.; Yang, H.-Y.; Gao, L.; Bai, M.-Z.; Fu, W.-K.; Huang, C.-F.; Mi, N.-N.; Ma, H.-D.; Lu, Y.-W.; Jiang, N.-Z.; et al. Lanatoside C Decelerates Proliferation and Induces Apoptosis through Inhibition of STAT3 and ROS-Mediated Mitochondrial Membrane Potential Transformation in Cholangiocarcinoma. Front. Pharmacol. 2023, 14, 1098915. [Google Scholar] [CrossRef]
  225. Li, X.-X.; Wang, D.-Q.; Sui, C.-G.; Meng, F.-D.; Sun, S.-L.; Zheng, J.; Jiang, Y.-H. Oleandrin Induces Apoptosis via Activating Endoplasmic Reticulum Stress in Breast Cancer Cells. Biomed. Pharmacother. 2020, 124, 109852. [Google Scholar] [CrossRef]
  226. Wang, L.; Cai, W.; Han, B.; Zhang, J.; Yu, B.; Chen, M. Ouabain Exhibited Strong Anticancer Effects in Melanoma Cells via Induction of Apoptosis, G2/M Phase Arrest, and Migration Inhibition. OncoTargets Ther. 2021, 14, 1261–1273. [Google Scholar] [CrossRef] [PubMed]
  227. Cheng, Y.; Wang, G.; Zhao, L.; Dai, S.; Han, J.; Hu, X.; Zhou, C.; Wang, F.; Ma, H.; Li, B.; et al. Periplocymarin Induced Colorectal Cancer Cells Apoptosis Via Impairing PI3K/AKT Pathway. Front. Oncol. 2021, 11, 753598. [Google Scholar] [CrossRef] [PubMed]
  228. Yang, Y.; Liu, Y.; Zhang, Y.; Ji, W.; Wang, L.; Lee, S.C. Periplogenin Activates ROS-ER Stress Pathway to Trigger Apoptosis via BIP-eIF2α- CHOP and IRE1α-ASK1-JNK Signaling Routes. Anticancer Agents Med. Chem. 2021, 21, 61–70. [Google Scholar] [CrossRef] [PubMed]
  229. Tian, X.; Gu, L.; Zeng, F.; Liu, X.; Zhou, Y.; Dou, Y.; Han, J.; Zhao, Y.; Zhang, Y.; Luo, Q.; et al. Strophanthidin Induces Apoptosis of Human Lung Adenocarcinoma Cells by Promoting TRAIL-DR5 Signaling. Molecules 2024, 29, 877. [Google Scholar] [CrossRef]
  230. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular Mechanisms and Health Implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef]
  231. Dixon, S.J.; Olzmann, J.A. The Cell Biology of Ferroptosis. Nat. Rev. Mol. Cell Biol. 2024, 25, 424–442. [Google Scholar] [CrossRef]
  232. Zhou, Q.; Meng, Y.; Li, D.; Yao, L.; Le, J.; Liu, Y.; Sun, Y.; Zeng, F.; Chen, X.; Deng, G. Ferroptosis in Cancer: From Molecular Mechanisms to Therapeutic Strategies. Signal Transduct. Target. Ther. 2024, 9, 1–30. [Google Scholar] [CrossRef]
  233. Tan, Z.; Huang, H.; Sun, W.; Li, Y.; Jia, Y. Current Progress of Ferroptosis Study in Ovarian Cancer. Front. Mol. Biosci. 2022, 9, 966007. [Google Scholar] [CrossRef]
  234. Chen, K.; Li, A.; Wang, J.; Li, D.; Wang, X.; Liu, C.; Wang, Z. Arenobufagin Causes Ferroptosis in Human Gastric Cancer Cells by Increasing Rev-Erbα Expression. J. Tradit. Complement. Med. 2023, 13, 72–80. [Google Scholar] [CrossRef]
  235. Long, J.; Wang, W.; Chu, J.; Li, Y.; Wang, M.; Su, J.; Yang, Y.; Wang, G.; Li, Q.; Cheng, H. Overexpression of Nrf2 Reverses Ferroptosis Induced by Arenobufagin in Gastric Cancer. Toxicol. Appl. Pharmacol. 2024, 484, 116842. [Google Scholar] [CrossRef]
  236. Wu, S.; Wu, X.; Wang, Q.; Chen, Z.; Li, L.; Chen, H.; Qi, H. Bufalin Induces Ferroptosis by Modulating the 2,4-Dienoyl-CoA Reductase (DECR1)-SLC7A11 Axis in Breast Cancer. Phytomedicine Int. J. Phytother. Phytopharm. 2024, 135, 156130. [Google Scholar] [CrossRef] [PubMed]
  237. Yu, P.; Zhang, X.; Liu, N.; Tang, L.; Peng, C.; Chen, X. Pyroptosis: Mechanisms and Diseases. Signal Transduct. Target. Ther. 2021, 6, 1–21. [Google Scholar] [CrossRef] [PubMed]
  238. Olona, A.; Hateley, C.; Guerrero, A.; Ko, J.-H.; Johnson, M.R.; Anand, P.K.; Thomas, D.; Gil, J.; Behmoaras, J. Cardiac Glycosides Cause Cytotoxicity in Human Macrophages and Ameliorate White Adipose Tissue Homeostasis. Br. J. Pharmacol. 2022, 179, 1874–1886. [Google Scholar] [CrossRef] [PubMed]
  239. Li, T.; Zhang, Y.; Li, H.; Zhang, H.; Xie, J.; Li, Z.; Zhang, K.; Yu, Y.; Mei, L. Bufalin CaCO3 Nanoparticles Triggered Pyroptosis through Calcium Overload via Na+/Ca2+ Exchanger Reverse for Cancer Immunotherapy. Nano Lett. 2024, 24, 12691–12700. [Google Scholar] [CrossRef]
  240. Gupta, G.; Afzal, M.; Moglad, E.; Goyal, A.; Almalki, W.H.; Goyal, K.; Rana, M.; Ali, H.; Rekha, A.; Kazmi, I.; et al. Parthanatos and Apoptosis: Unraveling Their Roles in Cancer Cell Death and Therapy Resistance. EXCLI J. 2025, 24, 351–380. [Google Scholar] [CrossRef]
  241. Zhou, M.; Boulos, J.C.; Klauck, S.M.; Efferth, T. The Cardiac Glycoside ZINC253504760 Induces Parthanatos-Type Cell Death and G2/M Arrest via Downregulation of MEK1/2 Phosphorylation in Leukemia Cells. Cell Biol. Toxicol. 2023, 39, 2971–2997. [Google Scholar] [CrossRef]
  242. Iba, T.; Helms, J.; Maier, C.L.; Ferrer, R.; Levy, J.H. Autophagy and Autophagic Cell Death in Sepsis: Friend or Foe? J. Intensive Care 2024, 12, 41. [Google Scholar] [CrossRef]
  243. Liu, S.; Yao, S.; Yang, H.; Liu, S.; Wang, Y. Autophagy: Regulator of Cell Death. Cell Death Dis. 2023, 14, 1–17. [Google Scholar] [CrossRef]
  244. Debnath, J.; Gammoh, N.; Ryan, K.M. Autophagy and Autophagy-Related Pathways in Cancer. Nat. Rev. Mol. Cell Biol. 2023, 24, 560–575. [Google Scholar] [CrossRef]
  245. Nguyen, T.H.; Nguyen, T.M.; Ngoc, D.T.M.; You, T.; Park, M.K.; Lee, C.H. Unraveling the Janus-Faced Role of Autophagy in Hepatocellular Carcinoma: Implications for Therapeutic Interventions. Int. J. Mol. Sci. 2023, 24, 16255. [Google Scholar] [CrossRef]
  246. Galluzzi, L.; Vitale, I.; Warren, S.; Adjemian, S.; Agostinis, P.; Martinez, A.B.; Chan, T.A.; Coukos, G.; Demaria, S.; Deutsch, E.; et al. Consensus Guidelines for the Definition, Detection and Interpretation of Immunogenic Cell Death. J. Immunother. Cancer 2020, 8, e000337. [Google Scholar] [CrossRef] [PubMed]
  247. Kroemer, G.; Galassi, C.; Zitvogel, L.; Galluzzi, L. Immunogenic Cell Stress and Death. Nat. Immunol. 2022, 23, 487–500. [Google Scholar] [CrossRef] [PubMed]
  248. Ahmed, A.; Tait, S.W.G. Targeting Immunogenic Cell Death in Cancer. Mol. Oncol. 2020, 14, 2994–3006. [Google Scholar] [CrossRef] [PubMed]
  249. Galluzzi, L.; Guilbaud, E.; Schmidt, D.; Kroemer, G.; Marincola, F.M. Targeting Immunogenic Cell Stress and Death for Cancer Therapy. Nat. Rev. Drug Discov. 2024, 23, 445–460. [Google Scholar] [CrossRef]
  250. 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]
  251. Sukkurwala, A.Q.; Adjemian, S.; Senovilla, L.; Michaud, M.; Spaggiari, S.; Vacchelli, E.; Baracco, E.E.; Galluzzi, L.; Zitvogel, L.; Kepp, O.; et al. Screening of Novel Immunogenic Cell Death Inducers within the NCI Mechanistic Diversity Set. Oncoimmunology 2014, 3, e28473. [Google Scholar] [CrossRef]
  252. Xiang, Y.; Chen, L.; Li, L.; Huang, Y. Restoration and Enhancement of Immunogenic Cell Death of Cisplatin by Coadministration with Digoxin and Conjugation to HPMA Copolymer. ACS Appl. Mater. Interfaces 2020, 12, 1606–1616. [Google Scholar] [CrossRef]
  253. Glaviano, A.; Singh, S.K.; Lee, E.H.C.; Okina, E.; Lam, H.Y.; Carbone, D.; Reddy, E.P.; O’Connor, M.J.; Koff, A.; Singh, G.; et al. Cell Cycle Dysregulation in Cancer. Pharmacol. Rev. 2025, 77, 100030. [Google Scholar] [CrossRef]
  254. Mhaidly, N.; Barake, N.; Trelcat, A.; Journe, F.; Saussez, S.; Descamps, G. Bufalin Suppresses Head and Neck Cancer Development by Modulating Immune Responses and Targeting the β-Catenin Signaling Pathway. Cancers 2024, 16, 2739. [Google Scholar] [CrossRef]
  255. Wang, Z.; Liu, F.; Huang, C.; Zhang, J.; Wu, J. Bufalin Inhibits Epithelial-Mesenchymal Transition and Increases Radiosensitivity of Non-Small Cell Lung Cancer via Inhibition of the Src Signaling. J. Thorac. Dis. 2023, 15, 123–134. [Google Scholar] [CrossRef]
  256. Li, J.; Ma, R.; Lv, J.-L.; Ren, Y.-S.; Tan, Y.-J.; Wang, H.-M.; Wang, Z.-E.; Wang, B.-S.; Yu, J.-N.; Wang, Y.-L.; et al. Telocinobufagin, a PLK1 Suppressor That Inhibits Tumor Growth and Metastasis by Modulating CDC25c and CTCF in HNSCC Cells. Phytomedicine 2024, 127, 155440. [Google Scholar] [CrossRef] [PubMed]
  257. Cavalcanti, B.C.; Soares, B.M.; Barreto, F.S.; Magalhães, H.I.F.; Ferreira, J.R.d.O.; Almeida, A.T.A.d.; Araújo Beserra Filho, J.I.; Silva, J.; Dos Santos, H.S.; Marinho, E.S.; et al. Hellebrigenin Triggers Death of Promyelocytic Leukemia Cells by Non-Genotoxic Ways. Toxicon Off. J. Int. Soc. Toxinology 2024, 238, 107591. [Google Scholar] [CrossRef] [PubMed]
  258. Zhang, X.; Yao, Z.; Xue, Z.; Wang, S.; Liu, X.; Hu, Y.; Zhang, Y.; Wang, J.; Li, X.; Chen, A. Resibufogenin Targets the ATP1A1 Signaling Cascade to Induce G2/M Phase Arrest and Inhibit Invasion in Glioma. Front. Pharmacol. 2022, 13, 855626. [Google Scholar] [CrossRef] [PubMed]
  259. Wang, Y.; Hou, Y.; Hou, L.; Wang, W.; Li, K.; Zhang, Z.; Du, B.; Kong, D. Digoxin Exerts Anticancer Activity on Human Nonsmall Cell Lung Cancer Cells by Blocking PI3K/Akt Pathway. Biosci. Rep. 2021, 41, BSR20211056. [Google Scholar] [CrossRef]
  260. Zhuang, Y.; Liu, K.; He, Q.; Gu, X.; Jiang, C.; Wu, J. Hypoxia Signaling in Cancer: Implications for Therapeutic Interventions. MedComm 2023, 4, e203. [Google Scholar] [CrossRef]
  261. Ortmann, B.M. Hypoxia-Inducible Factor in Cancer: From Pathway Regulation to Therapeutic Opportunity. BMJ Oncol. 2024, 3. [Google Scholar] [CrossRef]
  262. Han, Y.; Zhu, B.; Meng, S. Endothelial Cell in Tumor Angiogenesis: Origins, Mechanisms, and Therapeutic Implication. Genes Dis. 2025, 101611. [Google Scholar] [CrossRef]
  263. Lorenc, P.; Sikorska, A.; Molenda, S.; Guzniczak, N.; Dams-Kozlowska, H.; Florczak, A. Physiological and Tumor-Associated Angiogenesis: Key Factors and Therapy Targeting VEGF/VEGFR Pathway. Biomed. Pharmacother. 2024, 180, 117585. [Google Scholar] [CrossRef]
  264. Lin, J.; Denmeade, S.; Carducci, M.A. HIF-1alpha and Calcium Signaling as Targets for Treatment of Prostate Cancer by Cardiac Glycosides. Curr. Cancer Drug Targets 2009, 9, 881–887. [Google Scholar] [CrossRef]
  265. Fang, K.; Zhan, Y.; Zhu, R.; Wang, Y.; Wu, C.; Sun, M.; Qiu, Y.; Yuan, Z.; Liang, X.; Yin, P.; et al. Bufalin Suppresses Tumour Microenvironment-Mediated Angiogenesis by Inhibiting the STAT3 Signalling Pathway. J. Transl. Med. 2021, 19, 383. [Google Scholar] [CrossRef]
  266. Li, Y.; Zhou, L.; Sun, K.; Guo, R.; Li, Z.; Wen, Q.; Fu, G.; Yang, S. Integrated Network Pharmacology, Proteomics, Molecular Docking, and Experiments in Vivo and in Vitro to Explore the Efficacy and Potential Mechanism of Bufalin against Hepatocellular Carcinoma Angiogenesis. J. Ethnopharmacol. 2025, 345, 119589. [Google Scholar] [CrossRef] [PubMed]
  267. Zeng, R.; Zhou, R.; Zhen, L.; Lan, J.; Li, Z.; Gu, D.; Nie, W.; Shen, Y.; Zhang, M.; Zhang, T.; et al. Tumor-Targeted Nanosystem with Hypoxia Inducible Factor 1α Inhibition for Synergistic Chemo-Photodynamic Therapy against Hypoxic Tumor. Colloids Surf. B Biointerfaces 2025, 248, 114456. [Google Scholar] [CrossRef] [PubMed]
  268. Weng, L.; Zhao, M.; Chen, Z.; Zhu, L. Hypoxia-Targeted Responsive Delivery of Doxorubicin and Digoxin for Synergistic Treatment of Triple-Negative Breast Cancer. Mol. Pharm. 2025, 22, 2142–2158. [Google Scholar] [CrossRef] [PubMed]
  269. Castaneda, M.; den Hollander, P.; Kuburich, N.A.; Rosen, J.M.; Mani, S.A. Mechanisms of Cancer Metastasis. Semin. Cancer Biol. 2022, 87, 17–31. [Google Scholar] [CrossRef]
  270. Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular Principles of Metastasis: A Hallmark of Cancer Revisited. Signal Transduct. Target. Ther. 2020, 5, 28. [Google Scholar] [CrossRef]
  271. Wang, M.; Hu, S.; Yang, J.; Yuan, L.; Han, L.; Liang, F.; Zhang, F.; Zhao, H.; Liu, Y.; Gao, N. Arenobufagin Inhibits Lung Metastasis of Colorectal Cancer by Targeting C-MYC/Nrf2 Axis. Phytomedicine 2024, 127, 155391. [Google Scholar] [CrossRef]
  272. Ding, L.; Yang, Y.; Lu, Q.; Qu, D.; Chandrakesan, P.; Feng, H.; Chen, H.; Chen, X.; Liao, Z.; Du, J.; et al. Bufalin Inhibits Tumorigenesis, Stemness, and Epithelial-Mesenchymal Transition in Colorectal Cancer through a C-Kit/Slug Signaling Axis. Int. J. Mol. Sci. 2022, 23, 13354. [Google Scholar] [CrossRef]
  273. Wang, H.; Chen, J.; Li, S.; Yang, J.; Tang, D.; Wu, W.; Yu, K.; Cao, Y.; Xu, K.; Yin, P.; et al. Bufalin Reverses Cancer-Associated Fibroblast-Mediated Colorectal Cancer Metastasis by Inhibiting the STAT3 Signaling Pathway. Apoptosis Int. J. Program. Cell Death 2023, 28, 594–606. [Google Scholar] [CrossRef]
  274. Hou, R.; Liu, X.; Yang, H.; Deng, S.; Cheng, C.; Liu, J.; Li, Y.; Zhang, Y.; Jiang, J.; Zhu, Z.; et al. Chemically Synthesized Cinobufagin Suppresses Nasopharyngeal Carcinoma Metastasis by Inducing ENKUR to Stabilize P53 Expression. Cancer Lett. 2022, 531, 57–70. [Google Scholar] [CrossRef]
  275. Zhu, Z.; Wang, H.; Qian, X.; Xue, M.; Sun, A.; Yin, Y.; Tang, J.; Zhang, J. Inhibitory Impact Of Cinobufagin In Triple-Negative Breast Cancer Metastasis: Involvements Of Macrophage Reprogramming Through Upregulated MME and Inactivated FAK/STAT3 Signaling. Clin. Breast Cancer 2024, 24, e244–e257.e1. [Google Scholar] [CrossRef]
  276. Li, W.; Pei, S.; Zhang, X.; Qi, D.; Zhang, W.; Dou, Y.; Yang, R.; Yao, X.; Zhang, Z.; Xie, S.; et al. Cinobufotalin Inhibits the Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma Cells through down-Regulate β-Catenin in Vitro and in Vivo. Eur. J. Pharmacol. 2022, 922, 174886. [Google Scholar] [CrossRef] [PubMed]
  277. Karaś, K.; Sałkowska, A.; Dastych, J.; Bachorz, R.A.; Ratajewski, M. Cardiac Glycosides with Target at Direct and Indirect Interactions with Nuclear Receptors. Biomed. Pharmacother. 2020, 127, 110106. [Google Scholar] [CrossRef] [PubMed]
  278. Huh, J.R.; Leung, M.W.L.; Huang, P.; Ryan, D.A.; Krout, M.R.; Malapaka, R.R.V.; Chow, J.; Manel, N.; Ciofani, M.; Kim, S.V.; et al. Digoxin and Its Derivatives Suppress TH17 Cell Differentiation by Antagonizing RORγt Activity. Nature 2011, 472, 486–490. [Google Scholar] [CrossRef] [PubMed]
  279. Fujita-Sato, S.; Ito, S.; Isobe, T.; Ohyama, T.; Wakabayashi, K.; Morishita, K.; Ando, O.; Isono, F. Structural Basis of Digoxin That Antagonizes RORgamma t Receptor Activity and Suppresses Th17 Cell Differentiation and Interleukin (IL)-17 Production. J. Biol. Chem. 2011, 286, 31409–31417. [Google Scholar] [CrossRef]
  280. Cascão, R.; Vidal, B.; Raquel, H.; Neves-Costa, A.; Figueiredo, N.; Gupta, V.; Fonseca, J.E.; Moita, L.F. Effective Treatment of Rat Adjuvant-Induced Arthritis by Celastrol. Autoimmun. Rev. 2012, 11, 856–862. [Google Scholar] [CrossRef]
  281. Lee, J.; Baek, S.; Lee, J.; Lee, J.; Lee, D.-G.; Park, M.-K.; Cho, M.-L.; Park, S.-H.; Kwok, S.-K. Digoxin Ameliorates Autoimmune Arthritis via Suppression of Th17 Differentiation. Int. Immunopharmacol. 2015, 26, 103–111. [Google Scholar] [CrossRef]
  282. Titus, H.E.; Xu, H.; Robinson, A.P.; Patel, P.A.; Chen, Y.; Fantini, D.; Eaton, V.; Karl, M.; Garrison, E.D.; Rose, I.V.L.; et al. Repurposing the Cardiac Glycoside Digoxin to Stimulate Myelin Regeneration in Chemically-Induced and Immune-Mediated Mouse Models of Multiple Sclerosis. Glia 2022, 70, 1950–1970. [Google Scholar] [CrossRef]
  283. Tani, S.; Takano, R.; Tamura, S.; Oishi, S.; Iwaizumi, M.; Hamaya, Y.; Takagaki, K.; Nagata, T.; Seto, S.; Horii, T.; et al. Digoxin Attenuates Murine Experimental Colitis by Downregulating Th17-Related Cytokines. Inflamm. Bowel Dis. 2017, 23, 728–738. [Google Scholar] [CrossRef]
  284. Saeed, H.; Mateen, S.; Moin, S.; Khan, A.Q.; Owais, M. Cardiac Glycoside Digoxin Ameliorates Pro-Inflammatory Cytokines in PBMCs of Rheumatoid Arthritis Patients in Vitro. Int. Immunopharmacol. 2020, 82, 106331. [Google Scholar] [CrossRef]
  285. Karaś, K.; Sałkowska, A.; Sobalska-Kwapis, M.; Walczak-Drzewiecka, A.; Strapagiel, D.; Dastych, J.; Bachorz, R.A.; Ratajewski, M. Digoxin, an Overlooked Agonist of RORγ/RORγT. Front. Pharmacol. 2018, 9, 1460. [Google Scholar] [CrossRef]
  286. Karwaciak, I.; Pastwińska, J.; Sałkowska, A.; Bachorz, R.A.; Ratajewski, M. Evaluation of the Activity of Cardiac Glycosides on RORγ and RORγT Nuclear Receptors. Arch. Biochem. Biophys. 2024, 759, 110085. [Google Scholar] [CrossRef] [PubMed]
  287. Li, I.-H.; Pan, K.-T.; Wang, W.-M.; Chien, W.-C.; Shih, J.-H.; Kao, L.-T. Digoxin Use and Following Risk of Psoriasis: A Population-Based Cohort Study in Taiwan. J. Dermatol. 2020, 47, 458–463. [Google Scholar] [CrossRef] [PubMed]
  288. Zhao, Y.-Z.; Wang, Y.-L.; Yu, Y. Immunoenhancement Effect of Cinobufagin on Macrophages and the Cyclophosphamide-Induced Immunosuppression Mouse Model. Int. Immunopharmacol. 2024, 131, 111885. [Google Scholar] [CrossRef] [PubMed]
  289. Tang, Y.; Luo, J.; Qin, L.; Tang, C.; Qiu, C.; Li, J. Network Pharmacology and Molecular Docking-Based Screening of Immunotherapeutic Targets for HuaChanSu Against Breast Cancer. Mol. Biotechnol. 2024, 1–11. [Google Scholar] [CrossRef]
  290. Yuan, B.; He, J.; Kisoh, K.; Hayashi, H.; Tanaka, S.; Si, N.; Zhao, H.-Y.; Hirano, T.; Bian, B.; Takagi, N. Effects of Active Bufadienolide Compounds on Human Cancer Cells and CD4+CD25+Foxp3+ Regulatory T Cells in Mitogen-Activated Human Peripheral Blood Mononuclear Cells. Oncol. Rep. 2016, 36, 1377–1384. [Google Scholar] [CrossRef]
  291. Zhang, Y.; Shi, X.; Han, J.; Peng, W.; Fang, Z.; Zhou, Y.; Xu, X.; Lin, J.; Xiao, F.; Zhao, L.; et al. Convallatoxin Promotes M2 Macrophage Polarization to Attenuate Atherosclerosis Through PPARγ-Integrin Avβ5 Signaling Pathway. Drug Des. Devel. Ther. 2021, 15, 803–812. [Google Scholar] [CrossRef]
  292. Samolej, J.; White, I.J.; Strang, B.L.; Mercer, J. Cardiac Glycosides Inhibit Early and Late Vaccinia Virus Protein Expression. J. Gen. Virol. 2024, 105, 001971. [Google Scholar] [CrossRef]
  293. 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]
  294. Yang, C.-W.; Chang, H.-Y.; Lee, Y.-Z.; Hsu, H.-Y.; Lee, S.-J. The Cardenolide Ouabain Suppresses Coronaviral Replication via Augmenting a Na(+)/K(+)-ATPase-Dependent PI3K_PDK1 Axis Signaling. Toxicol. Appl. Pharmacol. 2018, 356, 90–97. [Google Scholar] [CrossRef]
  295. Mandal, D.; Feng, Z.; Stoltzfus, C.M. Excessive RNA Splicing and Inhibition of HIV-1 Replication Induced by Modified U1 Small Nuclear RNAs. J. Virol. 2010, 84, 12790–12800. [Google Scholar] [CrossRef]
  296. Škubník, J.; Bejček, J.; Pavlíčková, V.S.; Rimpelová, S. Repurposing Cardiac Glycosides: Drugs for Heart Failure Surmounting Viruses. Molecules 2021, 26, 5627. [Google Scholar] [CrossRef] [PubMed]
  297. Jahanshahi, S.; Ouyang, H.; Ahmed, C.; Zahedi Amiri, A.; Dahal, S.; Mao, Y.-Q.; Van Ommen, D.A.J.; Malty, R.; Duan, W.; Been, T.; et al. Broad Spectrum Post-Entry Inhibitors of Coronavirus Replication: Cardiotonic Steroids and Monensin. Virology 2024, 589, 109915. [Google Scholar] [CrossRef] [PubMed]
  298. Pacheco, B.; Fernández-Oliva, A.; García-Serradilla, M.; Risco, C. Digoxin Is a Potent Inhibitor of Bunyamwera Virus Infection in Cell Culture. J. Gen. Virol. 2023, 104. [Google Scholar] [CrossRef] [PubMed]
  299. Wu, S.; Wang, S.; Lin, X.; Yang, S.; Ba, X.; Xiong, D.; Xiao, L.; Li, R. Lanatoside C Inhibits Herpes Simplex Virus 1 Replication by Regulating NRF2 Distribution within Cells. Phytomedicine 2024, 124, 155308. [Google Scholar] [CrossRef]
  300. Qayed, W.S.; Ferreira, R.S.; Silva, J.R.A. In Silico Study towards Repositioning of FDA-Approved Drug Candidates for Anticoronaviral Therapy: Molecular Docking, Molecular Dynamics and Binding Free Energy Calculations. Molecules 2022, 27, 5988. [Google Scholar] [CrossRef]
  301. Carvalho, D.C.M.; Dunn, T.; Campos, R.K.; Tierney, J.A.; Onyoni, F.; Cavalcante-Silva, L.H.A.; Pena, L.J.; Rodrigues-Mascarenhas, S.; Wu, P.; Weaver, S.C. Antiviral and Immunomodulatory Effects of Ouabain against Congenital Zika Syndrome Model. Mol. Ther. J. Am. Soc. Gene Ther. 2025, 33, 465–470. [Google Scholar] [CrossRef]
  302. Wang, D.; Liu, J.; Zhu, Q.; Wei, X.; Zhang, X.; Chen, Q.; Zhao, Y.; Tang, H.; Xu, W. Ouabain Ameliorates Alzheimer’s Disease-Associated Neuropathology and Cognitive Impairment in FAD4T Mice. Nutrients 2024, 16, 3558. [Google Scholar] [CrossRef]
  303. Song, H.-L.; Demirev, A.V.; Kim, N.-Y.; Kim, D.-H.; Yoon, S.-Y. Ouabain Activates Transcription Factor EB and Exerts Neuroprotection in Models of Alzheimer’s Disease. Mol. Cell. Neurosci. 2019, 95, 13–24. [Google Scholar] [CrossRef]
  304. Erdogan, M.A.; Kirazlar, M.; Yigitturk, G.; Erbas, O. Digoxin Exhibits Neuroprotective Properties in a Rat Model of Dementia. Neurochem. Res. 2022, 47, 1290–1298. [Google Scholar] [CrossRef]
  305. Nguyen, L.D.; Wei, Z.; Silva, M.C.; Barberán-Soler, S.; Zhang, J.; Rabinovsky, R.; Muratore, C.R.; Stricker, J.M.S.; Hortman, C.; Young-Pearse, T.L.; et al. Small Molecule Regulators of microRNAs Identified by High-Throughput Screen Coupled with High-Throughput Sequencing. Nat. Commun. 2023, 14, 7575. [Google Scholar] [CrossRef]
  306. Valvassori, S.S.; Aguiar-Geraldo, J.M.; Possamai-Della, T.; da-Rosa, D.D.; Peper-Nascimento, J.; Cararo, J.H.; Quevedo, J. Depressive-like Behavior Accompanies Neuroinflammation in an Animal Model of Bipolar Disorder Symptoms Induced by Ouabain. Pharmacol. Biochem. Behav. 2022, 219, 173434. [Google Scholar] [CrossRef] [PubMed]
  307. Valvassori, S.S.; Dal-Pont, G.C.; Varela, R.B.; Resende, W.R.; Gava, F.F.; Mina, F.G.; Budni, J.; Quevedo, J. Ouabain Induces Memory Impairment and Alter the BDNF Signaling Pathway in an Animal Model of Bipolar Disorder: Cognitive and Neurochemical Alterations in BD Model. J. Affect. Disord. 2021, 282, 1195–1202. [Google Scholar] [CrossRef] [PubMed]
  308. Castro de Jesus, L.; Gonçalves-de-Albuquerque, C.F.; Burth, P. Onset of Bipolar Disorder by COVID-19: The Roles of Endogenous Ouabain and the Na,K-ATPase. J. Psychiatr. Res. 2024, 179, 60–68. [Google Scholar] [CrossRef] [PubMed]
  309. Tsyvunin, V.; Shtrygol, S.; Mishchenko, M.; Lytkin, D.; Taran, A.; Shtrygol, D.; Gorbach, T. Effect of Digoxin, Sodium Valproate, and Celecoxib on the Cerebral Cyclooxygenase Pathway and Neuron-Specific Enolase under the Pentylenetetrazole-Induced Kindling in Mice. Ceska Slov. Farm. Cas. Ceske Farm. Spolecnosti Slov. Farm. Spolecnosti 2023, 72, 172–183. [Google Scholar] [CrossRef]
  310. Sibarov, D.A.; Zhuravleva, Z.D.; Ilina, M.A.; Boikov, S.I.; Stepanenko, Y.D.; Karelina, T.V.; Antonov, S.M. Unveiling the Role of Cholesterol in Subnanomolar Ouabain Rescue of Cortical Neurons from Calcium Overload Caused by Excitotoxic Insults. Cells 2023, 12, 2011. [Google Scholar] [CrossRef]
  311. Hashimoto, J.; Fujita, E.; Tanimoto, K.; Kondo, S.; Matsumoto-Miyai, K. Effects of Cardiac Glycoside Digoxin on Dendritic Spines and Motor Learning Performance in Mice. Neuroscience 2024, 541, 77–90. [Google Scholar] [CrossRef]
  312. Laudisio, A.; Marzetti, E.; Pagano, F.; Cocchi, A.; Bernabei, R.; Zuccalà, G. Digoxin and Cognitive Performance in Patients with Heart Failure: A Cohort, Pharmacoepidemiological Survey. Drugs Aging 2009, 26, 103–112. [Google Scholar] [CrossRef]
  313. Cao, J.; Yao, D.; Li, R.; Guo, X.; Hao, J.; Xie, M.; Li, J.; Pan, D.; Luo, X.; Yu, Z.; et al. Digoxin Ameliorates Glymphatic Transport and Cognitive Impairment in a Mouse Model of Chronic Cerebral Hypoperfusion. Neurosci. Bull. 2022, 38, 181–199. [Google Scholar] [CrossRef]
  314. Li, T.; Song, X.; Chen, J.; Li, Y.; Lin, J.; Li, P.; Yu, S.; Durojaye, O.A.; Yang, F.; Liu, X.; et al. Kupffer Cell-Derived IL6 Promotes Hepatocellular Carcinoma Metastasis Via the JAK1-ACAP4 Pathway. Int. J. Biol. Sci. 2025, 21, 285–305. [Google Scholar] [CrossRef]
  315. Pavithran, H.; Kumavath, R.; Ghosh, P. Transcriptome Profiling of Cardiac Glycoside Treatment Reveals EGR1 and Downstream Proteins of MAPK/ERK Signaling Pathway in Human Breast Cancer Cells. Int. J. Mol. Sci. 2023, 24, 15922. [Google Scholar] [CrossRef]
  316. Meng, Q.; Liu, K.; Liu, Z.; Liu, J.; Tian, Z.; Qin, S.; Wei, J.; Cheng, L. Digoxin Protects against Intervertebral Disc Degeneration via TNF/NF-κB and LRP4 Signaling. Front. Immunol. 2023, 14, 1251517. [Google Scholar] [CrossRef] [PubMed]
  317. Wang, K.; Ding, X.; Jiang, N.; Zeng, C.; Wu, J.; Cai, X.-Y.; Hettinghouse, A.; Khleborodova, A.; Lei, Z.-N.; Chen, Z.-S.; et al. Digoxin Targets Low Density Lipoprotein Receptor-Related Protein 4 and Protects against Osteoarthritis. Ann. Rheum. Dis. 2022, 81, 544–555. [Google Scholar] [CrossRef] [PubMed]
  318. Tian, D.; Tang, J.; Geng, X.; Li, Q.; Wang, F.; Zhao, H.; Narla, G.; Yao, X.; Zhang, Y. Targeting UHRF1-Dependent DNA Repair Selectively Sensitizes KRAS Mutant Lung Cancer to Chemotherapy. Cancer Lett. 2020, 493, 80–90. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure and diversity of cardiac glycosides. (a) Core steroidal structure common to all CGs, showing the carbon numbering across the four fused rings (A, B, C, and D). (b) Three-dimensional representation of the steroidal nucleus, highlighting the characteristic U-shaped conformation of the pharmacophore. (c) Chemical structures of representative cardenolides. (d) Chemical structures of representative bufadienolides.
Figure 1. Chemical structure and diversity of cardiac glycosides. (a) Core steroidal structure common to all CGs, showing the carbon numbering across the four fused rings (A, B, C, and D). (b) Three-dimensional representation of the steroidal nucleus, highlighting the characteristic U-shaped conformation of the pharmacophore. (c) Chemical structures of representative cardenolides. (d) Chemical structures of representative bufadienolides.
Biomolecules 15 00885 g001
Figure 2. Diversity and origin of naturally occurring cardiac glycosides (CGs). The top panel illustrates the structural differences between cardenolides and bufadienolides. The lower panels depict various plant and animal species known to produce CGs—cardenolide-producing species are shown on the left, and bufadienolide-producing species on the right. For each organism, both the scientific name and the corresponding type(s) of CGs they produce are indicated.
Figure 2. Diversity and origin of naturally occurring cardiac glycosides (CGs). The top panel illustrates the structural differences between cardenolides and bufadienolides. The lower panels depict various plant and animal species known to produce CGs—cardenolide-producing species are shown on the left, and bufadienolide-producing species on the right. For each organism, both the scientific name and the corresponding type(s) of CGs they produce are indicated.
Biomolecules 15 00885 g002
Figure 5. Signaling pathways activated by the binding of CGs to NKA. Arrow colors correspond to different signaling pathways, as detailed in the text.
Figure 5. Signaling pathways activated by the binding of CGs to NKA. Arrow colors correspond to different signaling pathways, as detailed in the text.
Biomolecules 15 00885 g005
Figure 6. Influence of cardiac glycosides on physiological and pathological processes. Red arrows indicate deleterious effects on target cells. Green arrows represent physiological (beneficial) effects. Yellow arrows indicate context-dependent effects, which may be either beneficial or harmful depending on dosage and target cell type, as discussed in the text.
Figure 6. Influence of cardiac glycosides on physiological and pathological processes. Red arrows indicate deleterious effects on target cells. Green arrows represent physiological (beneficial) effects. Yellow arrows indicate context-dependent effects, which may be either beneficial or harmful depending on dosage and target cell type, as discussed in the text.
Biomolecules 15 00885 g006
Figure 7. CGs. Influence on key hallmarks of the epithelial phenotype. Color of arrows represent different signalling pathways.
Figure 7. CGs. Influence on key hallmarks of the epithelial phenotype. Color of arrows represent different signalling pathways.
Biomolecules 15 00885 g007
Table 1. Diversity of CGs’ impact on cancer types.
Table 1. Diversity of CGs’ impact on cancer types.
NameTarget/PathwayEffectTissueRef
Arenobufaginc-Jun N-terminal kinases (JNK)ApoptosisNasopharyngeal carcinoma[162]
MiR-149-5p/AEBP1FerroptosisGlioblastoma[163]
β-cateninEMTProstate[164]
p62-Keap1-Nrf2 AutophagyLiver[165]
IKKβ/NFκB MigrationLung[166]
Acetyl-bufalinCDK9/STAT3GrowthNon-small lung[55]
Acetyl-cinobufaginSTAT3Proliferation, migration, EMTBreast[167]
BufalinSRC-3/c-MycMetastasisColon[168]
SRC-3/HIF-1αGlycolysisColon[169]
Ca2+/CaMKKβ/AMPK/Beclin1Apoptosis, autophagyOsteosarcoma[170]
CAMKK2/Wnt/β-cateninProliferation, metastasisBile ducts[171]
Hippo-YAPProliferationLung[172]
PIAS3/STAT3Proliferation, migration, invasionEsophagus[173]
BFAR/PI3K/AKT/mTORMetastasisStomach[102]
AK/STAT, Wnt/β-Catenin, mTOR, TRAIL/TRAIL-RProliferation, metastasisVarious[174]
BufotalinAKTApoptosisGlioblastoma[175]
CerberinPI3K/AKT/mTORApoptosisND[101]
CinobufaginPI3K/AKT, MAPK/ERKGrowthLung[176]
CinobufotalinUSP36/c-Myc axisProliferation, migration, invasionColon[177]
ConvallatoxinWnt/β-catenin Proliferation, migration, invasionBone[178]
DigitoxinNF-κB/ST6GAL1Proliferation, migrationLiver[179]
DigoxinHIF-1αGrowthND[108]
STAT3MigrationLung[180]
GamabufotalinTGF-β/periostin/PI3K/AKTMetastasisBones[181]
NAK(ATP1A3)-AQP4GrowthGlioblastoma[182]
Lanatoside CMAPK, Wnt, JAK-STAT, and PI3K/AKT/mTOR GrowthBreast, lung, liver[183]
TNF/IL-17 Proliferation, apoptosisProstate[184]
MalayosideMAPK-Nur77ApoptosisNon-small lung[185]
Odoroside AROS/JNK ProliferationLeukemia[186]
STAT-3 InvasionBreast[187]
OleandrinPERK/elF2α/ATF4/CHOPImmunogenic cell deathBreast[118,188]
OuabainAMPK-Src Autophagy, metabolism [189]
PeruvosideMAPK Wnt/β-catenin, PI3K/AKT/mTOR GrowthBreast, lung, and liver[117]
Src-EGFRGrowth, invasionLung[190]
PeriplogeninJAK2/3-STAT3GrowthEsophagus[191]
ResibufogeninPI3K/AKT aGrowth, migrationOvary[192]
ResibufogeninPI3K/AktViability, migration invasion,Bone marrow[193]
VEGFR2-(VEG)AngiogenesisBreast[194]
lncRNA LINC00597/hsa-miR-367-3p/TFRC FerroptosisLung[195]
TelocinobufaginLARP1-mTORMetastasisThyroid[196]
STAT3/PARP1ApoptosisLung[197]
αldiginosideJAK-STATApoptosisND[198]
Table 2. Variety of CGs inducing apoptosis in cancer cells through distinct targets and signaling mechanisms.
Table 2. Variety of CGs inducing apoptosis in cancer cells through distinct targets and signaling mechanisms.
CG NameTarget or PathwayCancer TypeRef
21-Benzylidene digoxin(−) EGFR/ERKHeLa cells[50]
ArenobufaginModulating claspin and JNK pathwayNasopharyngeal carcinoma cells[162]
Induces apoptosis and G2/M arrestA549 cells[211]
(−) PI3K/AKT/mTORPancreatic Cancer Cells[212]
BufalinCa2+/CaMKKβ/AMPK/Beclin1Osteosarcoma cells[170]
Annexin A2 and DRP1 regulationGlioma cells[213]
ROSNeuroblastoma[214]
UnspecifiedGlioma[215]
BufarenoginBax and ANT cooperationUnspecified[216]
BufotalinMitochondrial dysfunction via AKT signaling pathwayGlioblastoma cells[175]
Inhibiting the STAT3/EMT AxisTriple-negative breast cancer cells[217]
CinobufaginG9aNon-small-cell lung cancer A549 cells[218]
(−) β-catenin signalingAcute promyelocytic leukemia[219]
DNA damage response, G2/M checkpointUnspecified cancer cells[220]
UnspecifiedNasopharyngeal carcinoma cells[221]
ConvallatoxinJAK2/STAT3 and mTOR/STAT3Colorectal cancer[116]
DigitoxinHIF-1α and STAT3KRAS mutant human colon cancer cells[222]
HellebrigeninMAPK signaling and XIAP expressionOral cancer[223]
Lanatoside CTNF/IL-17 signaling pathwayHuman prostate cancer cells[184]
Inhibition of STAT3Cholangiocarcinoma[224]
MalayosideMAPK-Nur77 signalingHuman non-small lung cancer cells[185]
OleandrinROS-ER StressBreast cancer cells[225]
OuabainInduction of apoptosis, G2/M arrest, migration inhibitionMelanoma cells[226]
PeriplocymarinPI3K/AKT pathwayColorectal cancer cells[227]
PeriplogeninROS-ER stress Unspecified[228]
PeruvosideMAPK, Wnt/β-catenin, PI3K/AKT/mTORHuman cancers[117]
StrophanthidinPromoting TRAIL-DR5 signalingLung Adenocarcinoma[229]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ponce, A.; Flores-Maldonado, C.; Contreras, R.G. Cardiac Glycosides: From Natural Defense Molecules to Emerging Therapeutic Agents. Biomolecules 2025, 15, 885. https://doi.org/10.3390/biom15060885

AMA Style

Ponce A, Flores-Maldonado C, Contreras RG. Cardiac Glycosides: From Natural Defense Molecules to Emerging Therapeutic Agents. Biomolecules. 2025; 15(6):885. https://doi.org/10.3390/biom15060885

Chicago/Turabian Style

Ponce, Arturo, Catalina Flores-Maldonado, and Ruben G. Contreras. 2025. "Cardiac Glycosides: From Natural Defense Molecules to Emerging Therapeutic Agents" Biomolecules 15, no. 6: 885. https://doi.org/10.3390/biom15060885

APA Style

Ponce, A., Flores-Maldonado, C., & Contreras, R. G. (2025). Cardiac Glycosides: From Natural Defense Molecules to Emerging Therapeutic Agents. Biomolecules, 15(6), 885. https://doi.org/10.3390/biom15060885

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

Article Metrics

Back to TopTop