Next Article in Journal
PhosphoLipidome Alteration Induced by Clostridioides difficile Toxin B in Enteric Glial Cells
Next Article in Special Issue
Understanding Macrophage Interaction with Antimony-Doped Tin Oxide Plasmonic Nanoparticles
Previous Article in Journal
Peripherally Restricted CB1 Receptor Inverse Agonist JD5037 Treatment Exacerbates Liver Injury in MDR2-Deficient Mice
Previous Article in Special Issue
Green-Synthesized Silver and Selenium Nanoparticles Using Berberine: A Comparative Assessment of In Vitro Anticancer Potential on Human Hepatocellular Carcinoma Cell Line (HepG2)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Mechanisms of the Therapeutic Effect of Selenium Nanoparticles in Hepatocellular Carcinoma

by
Elena G. Varlamova
Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia
Cells 2024, 13(13), 1102; https://doi.org/10.3390/cells13131102
Submission received: 13 May 2024 / Revised: 10 June 2024 / Accepted: 21 June 2024 / Published: 26 June 2024

Abstract

:
This review describes and summarizes, for the first time, the molecular mechanisms of the cytotoxic effect of selenium nanoparticles of various origins on hepatocellular carcinoma cells. The text provides information from recent years indicating the regulation of various signaling pathways and endoplasmic reticulum stress by selenium nanoparticles; the pathways of cell death of liver cancer cells as a result of exposure to selenium nanoparticles are considered. Particular attention is paid to the participation of selenoproteins and selenium-containing thioredoxin reductases and glutathione peroxidases in these processes. Previously, there were no reviews that fully reflected the cytotoxic effects of selenium nanoparticles specifically in hepatocellular carcinoma, despite the fact that many reviews and experimental articles have been devoted to the causes of this disease and the molecular mechanisms of regulation of cytotoxic effects by other agents. The relevance of this review is primarily explained by the fact that despite the development of various drugs and approaches for the treatment and prevention of hepatocellular carcinoma, this disease is still the fourth leading cause of death in the world. For this reason, a complete understanding of the latest trends in the treatment of oncology of various etiologies, especially hepatocellular carcinoma, is extremely important.

Graphical Abstract

1. Introduction

Hepatocellular carcinoma (HCC) is the most common primary liver cancer and the fourth leading cause of death worldwide, the main cause of which is cirrhosis, regardless of its etiology; other common causes include carcinogens and genetic disorders [1,2]. Cancer development is very often characterized by the coevolution of cancer and immune cells [3,4]. HCC is not just a mixture of cells and extracellular matrix but several cell types that interact with each other and with surrounding tissues, creating a complex network of interactions. Multikinase inhibitors such as sorafenib, lenvatinib, regorafenib and cabozantinib, as well as the vascular endothelial growth factor (VEGF) inhibitor ramucirumab, are widely used clinically for the treatment of HCC [5,6,7,8]. In recent years, new therapeutic strategies have also been developed, such as immunosuppressive cancer therapy based on immune checkpoint inhibitors, with the combination of this method with the drug ramucirumab in advanced HCC showing better results compared with the widely used multikinase inhibitor sorafenib [9], while the combination of atezolizumab and bevacizumab is now positioned as first-line therapy for patients with advanced HCC. Increasingly, nanotechnology has recently been used to treat oncology of various etiologies; in particular, a lot of work has been devoted to the study of nanoparticles and nanocomplexes for the treatment of HCC, since they provide targeted delivery of drugs, increasing their therapeutic effectiveness. Among nanoparticles based on metals and nonmetals in HCC, the most widely used are gold nanoparticles, which have a high ability to bind to proteins and lipids on the surface of cancer cells [10]; iron nanoparticles, which are used in magnetic resonance analysis of HCC due to their interaction with external magnetic fields; and silver nanoparticles doped with curcumin, which have exhibited powerful antioxidant, anticarcinogenic, anti-inflammatory and antiangiogenic activity [11].
To date, sufficient research has been accumulated on the role of selenium nanoparticles (SeNPs) in the treatment of liver cancer. Such research is usually divided into two areas, namely the study of chemoprotective and chemosensitizing activities and the study of direct cytotoxic activity against liver cancer cells [12,13,14]. Almost always, treatment with SeNPs is accompanied by normalization of reactive oxygen species (ROS) and lipid peroxidation (LPO) levels; a decrease in the blood levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) enzymes; and an increase in the activity of enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), glutathione peroxidase (GPX) and glutathione S-transferase (GST) [15,16,17].
This review is devoted to describing the latest data regarding the molecular mechanisms of the cytotoxic effect of selenium-based nanoparticles towards HCC. For the first time, generalized information on the regulation of endoplasmic reticulum stress (ER-stress) and ER-resident selenoproteins activated in HCC by SeNPs is provided, and the putative functions of these selenoproteins in the progression or inhibition of HCC are described. Despite the fact that, recently, more and more information has appeared on the role of SeNPs in the treatment of HCC, no review has considered the molecular mechanisms of the regulation of hepatocarcinogenesis controlled by these nanoparticles and combined all the available information on their cytotoxicity in relation to HCC, which was the global goal of this review.

2. The “Therapeutic Window” of SeNPs Determines Their Hepatoprotective Functions

It is known that the antioxidant activity is one of the main functions of Se-containing compounds of organic and inorganic nature, including selenoproteins and Se-containing nanoparticles. Antioxidants can prevent the development of cancer by influencing the cell cycle, inflammation, proliferation, tumor invasiveness, apoptosis mechanisms, etc. [18]. On the other hand, ROS are essential for the adaptive response of cells, as well as their survival. In many ways, the answer to the question about the dangers and benefits of antioxidants lies in the used concentration range, depending on which they can behave as either pro- or antioxidants. It is known that Se can cause various toxic symptoms and has a narrow safe dosage range. In infectious, oncological and toxic diseases, the daily requirement for Se to protect the body increases from 50–100 μg to 600 μg/day per person, while the toxic dose is 900 μg/day per person [19].
Among all Se-containing agents, SeNPs have the least risk of toxicity, but the mechanisms they activate may differ depending on the cell type (normal or cancerous), the type of cancer cells, the dosage and the physicochemical properties of the nanoparticles themselves [20,21,22,23,24,25]. A number of studies have demonstrated that SeNPs can induce apoptotic death of cancer cells, for example, through the p53 and AKT pathways [26,27], by activating the mitochondria-mediated pathway [28,29], by inhibiting the EGFR (epidermal growth factor receptor)-mediated PI3K/AKT pathways and Ras/Raf/MEK/ERK and through activation of MAPK and the caspase-3 signaling pathway [30,31]. All these works highlight the anticancer potential of nanoparticles.
An alternative mechanism for the therapeutic properties of SeNPs is autophagic death of cancer cells, which can be triggered by enhancing signaling pathways associated with beclin-1 [32], suppressing the p62 protein [33,34,35] and by inhibiting the PI3K/AKT/mTOR pathway. On the other hand, there are studies indicating inhibition of autophagy by disrupting the function of lysosomes, alkalizing the lysosomal environment and blocking the late stage of autophagy [36,37].
However, it should be emphasized that excessive doses of SeNPs certainly have a cytotoxic effect [38,39]. Thus, in a carbon tetrachloride-induced inflammatory model in rats, high doses of SeNPs (10 mg/100 g) stabilized by a natural sulfated polysaccharide caused serious disorders associated with dysfunction of the system for excreting excess Se in the form of its methylated derivatives by the kidneys. This led to excess Se, which interacted with glutathione, leading to the generation of ROS and damage to liver tissue. In addition, an increase in biochemical indicators of LPO in the blood of experimental animals was observed, which indicates damage to the urinary tract [40].
In another study, when studying the toxicity of SeNPs at high (4 mg/kg/day), medium (0.5 mg/kg/day) and low (0.05 mg/kg/day) doses, it was found that high doses of SeNPs cause an increase in the relative weight of the liver, changes in biochemical parameters of plasma and urine creatinine, an increase in urine pH, etc. [41].
Thus, despite the presence of a number of significant advantages of SeNPs over other Se-containing forms, in each specific case, it is important to determine the “therapeutic window” to minimize the negative effects of excess SeNPs.

3. SeNPs as Inducers of Various Forms of Cell Death Using the Example of Liver Cancer Cells

3.1. SeNPs as an Inducer of Apoptosis in Liver Cancer Cells

It is currently believed that apoptosis is one of the most important mechanisms of chemoprevention and chemotherapy with Se compounds, including SeNPs, which cause apoptotic death of liver cancer cells, which has been demonstrated in a number of studies [27,42,43,44,45,46,47,48,49,50].
It has been shown that ultrasmall (about 5 nm) SeNPs nanolyzed with polyethylene glycol promote mitochondrial depolarization, which leads to apoptotic HepG2 cell death [43].
In a comparative analysis of nanoparticles doped with and without sorafenib, approximately 100 nm in size using the example of HepG2 cells, it was found that the most effective was the nanocomplex of Se and sorafenib, which, at a concentration of 0.5 μg/mL contributed to the induction of early stages of apoptosis after 24 h, whereas increasing the concentration of the nanocomplex by five times led to the induction of late stages of apoptosis. SeNPs of both types promoted an increase in the expression of a number of pro-apoptotic genes, including GADD34, BAK, BAX, PUMA, CASP-3 and CASP-4 [45].
It was also demonstrated that siRNA with polyethylenimine (PEI)-modified SeNPs enhanced apoptosis of HepG2 cells by reducing the expression of heat-shock protein HSP70 mRNA, which was accompanied by the inhibition of the protective function of these cells and subsequent apoptotic death. DNA fragmentation and nuclear condensation were observed, which are important biochemical signs of cellular apoptosis. Treatment of HepG2 cells with various concentrations of this nanocomplex significantly increased the activity of caspase-3 and the cleavage of DNA repair enzyme PARP in a dose-dependent manner [48].
Laminarin polysaccharide-decorated SeNPs (LP-SeNPs) with a diameter of 60 nm also exhibited cytotoxicity towards HepG2 cells. Thus, after their treatment with various concentrations of LP-SeNPs (10, 20 and 40 μM) for 24 h, average increases in the total rate of apoptosis of 17.4, 20.9 and 30.9%, respectively, were observed. These processes were accompanied by increases in BAX mRNA expression and caspase-9 cleavage and a decrease in BCL-2 levels. The obtained data most likely indicate the activation of mitochondrially mediated apoptosis by SeNPs [36].
SeNPs synthesized using hawthorn fruit extract (HE) as a reducing and stabilizing agent (HE-SeNPs) at concentrations of 5, 10 and 20 μg/mL caused early and late stages of apoptotic death of HepG2 cells [49]. In this case, the rate of apoptosis increased to 7.3, 9.7 and 19.2%, respectively; there was an increase in intracellular levels of ROS, a decrease in membrane potential, an increase in the level of caspase-9 and a decrease in the level of mRNA of anti-apoptotic protein BCL-2. Thus, He-SeNPs induced intracellular oxidative stress and mitochondrial dysfunction by initiating apoptosis of HepG2 cells through the mitochondrial pathway.
It was found that BFP (triple-helix β-glucan)-SeNPs were more effective than naked SeNPs, significantly reducing proliferation and disrupting the cell cycle and apoptosis of HepG2 cells [50]. Here, BFP not only acted as a stabilizer of SeNPs but also activated death receptor-mediated and mitochondrially mediated apoptosis pathways in liver cancer cells. However, BFP itself exhibited negligible cytotoxic activity against HepG2 cells, even when cells were treated with a dose of 200 μg/mL for 72 h. Naked SeNPs had a slightly better effect. But the BFP-SeNP nanocomplex showed significant synergistic effects, including the inhibition of cell proliferation through cell cycle arrest in the S phase, condensation of nuclear chromatin and severe nuclear shrinkage, an increase in ROS, a decrease in mitochondrial membrane potential and activation of the endogenous mitochondrial apoptosis pathway [50].

3.2. SeNPs as Inducers of Autophagy in Liver Cancer Cells

Recently, more and more evidence has emerged indicating the important role of SeNPs in the treatment of cancer and various infectious diseases through the regulation of autophagy, which is one of the most important intracellular processes of the degradation system [51].
Using the example of the above-mentioned laminarin-coated SeNPs (LP-SeNPs), it was demonstrated that exposure of HepG2 cells to this nanocomplex for 12 h led to the activation of light chain 3-II (LC3-II) and p62, important participants in the processes occurring during autophagy [52]. LC3 is a soluble protein weighing 17 kDa associated with microtubules, the cytosolic form of which (LC3-I) is conjugated with phosphatidylethanolamine to form LC3-II, which is recruited to autophagosomal membranes, that is, autophagy begins with the formation of a phagophore closely associated with LC3-II [52]. The first autophagy adapter discovered in mammals is p62 [53,54]. Therefore, biochemical methods based on the analysis of LC3-II and p62 are key in assessing the degree of autophagy progression in mammalian cells. These data suggest that LP-SeNPs induce the activation of early autophagy but block the late phase of autophagy. Since the damaged organelles could not be cleared, there was a decrease in the fusion of autophagosomes with lysosomes and a decrease in the enzymatic activity of lysosomes, which ultimately worsened the apoptosis of HepG2 cells.
To date, sufficient data have not yet been accumulated to convincingly prove the role of SeNPs in the regulation of autophagy processes in liver cancer cells; however, it has been shown that after 24 h and especially after 48 h, SeNPs doped with sorafenib (SeSo) are able to reduce the expression of mTOR, AKT and PI3K [45]. It is known that mammalian rapamycin kinase mTOR is a negative central regulator of the autophagic process; therefore, inhibition of the PI3K/AKT/mTOR pathway leads to the activation of autophagy, which is also important evidence of the death of HepG2 cells through autophagy after 24 and 48 h of treatment with SeNPs and SeSo nanoparticles.

4. Signaling Pathways Activated by SeNPs in HCC

The signaling mechanisms that SeNPs can trigger in liver cancer cells have not been sufficiently studied. However, there are already some general trends to consider with respect to some of the signaling pathways in which SeNPs are involved. Among them, it is worth highlighting the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR), Wnt/β-catenin and RAS/RAF/UPR (unfolded protein response) signaling pathways during endoplasmic reticulum stress.

4.1. Participation of SeNPs in the Regulation of ER Stress in HCC

Endoplasmic reticulum stress (ER stress) is a typical molecular pathophysiological process that underlies many human diseases, for the development of which disruption of protein folding is of great importance, which can lead to the accumulation of inactive or chemically aggressive proteins in the lumen of the ER [55].
When assessing the cytotoxic effect of “naked” SeNPs, SeNPs coated with sorafenib (SeSo), with the drug sorafenib (So) using the example of HepG2 cells, it was found that all agents are able to activate UPR signaling pathways when exacerbating ER stress [45]. In this study, it was found that after 24 h of treatment of HepG2 cells with So and SeSo, there was an increase in the expression of the mRNA of transcription factor ATF-4, which is a key marker of the PERK (protein kinase RNA-like endoplasmic reticulum kinase) signaling pathway of the UPR [56,57,58,59,60]. However, no increase in the expression of this transcription factor was observed after treatment of cells with SeNPs. On the contrary, after treatment of HepG2 cells with SeNPs and SeSo, a noticeable decrease in the spliced form of transcription factor XBP1 (XBP1s), which can be considered a key marker of the IRE1α signaling pathway of the UPR, was observed [61,62,63].
Since this work established that treatment of HepG2 cells with SeNPs, So and SeSo for 24 h promoted apoptotic death, this most likely occurred, among other things, as a result of prolonged ER stress, which was accompanied by the activation of not the adaptive but pro-apoptotic UPR signaling pathways. The work also shows the connection of SeNPs and SeSo nanoparticles with the dose-dependent generation of various calcium signals (single pulses and calcium oscillations). At the same time, an increase in the expression of caspase-4 was observed, which is activated under ER stress conditions, including through the Ca2+ signaling pathway [64,65,66]. It is known that during ER stress, conformational changes and/or oligomerization of pro-apoptotic proteins BAX and BAK occur, an increase in expression of which was also recorded in this work [45], which can lead to damage to calcium stores in the ER and the release of calcium into the cytosol [67,68]. This, in turn, activates cysteine protease m-calpain, which further cleaves procaspase-12 to caspase-12, leading to the activation of apoptosis [64,65,66]. Caspase-12 has been identified in rodents, and in humans, its role is presumably played by caspase-4 [69].
However, these experiments showed that SeNPs and SeSo are able to enhance the growth of pro-apoptotic markers CHOP, BIM, PUMA, GADD34, BAX and BAK mRNA expression and, at the same time, reduce the expression of anti-apoptotic gene BCL-2 [45]. It is well known that the activation of CHOP (CCAAT/enhancer-binding protein-homologous protein) occurs as a result of the activation of the UPR PERK signaling pathway described above [57,70]. Both anti-apoptotic gene BCL-2 and pro-apoptotic BIM and BAX have been shown to be regulated by CHOP during ER stress, which suppresses BCL-2 expression, activates BIM and promotes BAX translocation into mitochondria [71,72]. These events may cause damage to the outer mitochondrial membrane, a decrease in membrane potential and the release of cytochrome C, which, together with the APAF-1 protein (apoptotic protease activating factor-1), is involved in the formation of the apoptosome. Next, CASP-9 is activated, which binds and activates pro-CASP-3 to form effector CASP-3, which leads to apoptosis of HepG2 cells. These processes are shown schematically in Figure 1.
SeNPs activate the PERK signaling pathway UPR as a cell response to increasing ER stress. In this case, the expression of the key marker of this signaling pathway, ATF-4, increases, which serves as a trigger for increased expression of genes such as CHOP, GADD34, BAX and BAK, which ultimately leads to the discovery of mitochondrial membrane permeability, the release of cytochrome C into the cytoplasm, activation of caspase- 9, apoptosome formation, caspase-3 activation and cell apoptosis. These processes are also accompanied by an increase in the concentration of cytosolic calcium, the activation of caspase 4 and further activation of caspases 9 and 3, which also leads to apoptosis of cancer cells. SeNPs can be a source of ROS of various natures, which leads to the activation (phosphorylation) of p53, which, in turn, blocks the expression of anti-apoptotic gene BCL-2 and activates increased expression of various pro-apoptotic genes of the BCL family (BAX, BAK, PUMA, NOXA and BID), which ultimately activates mitochondrially mediated apoptosis. SeNPs are able to enhance apoptosis of HepG2 cells by reducing the expression of heat-shock protein HSP70 mRNA, which is accompanied by DNA fragmentation, nuclear condensation, caspase-3 activation and apoptosis.

4.2. Participation of SeNPs in Wnt/β-Catenin Signaling in HCC

To date, there is sufficient evidence that Wnt/β-catenin signaling is active in HCC, since in liver tumors, HCC cells and macrophages are new sources of the Wnt ligand (from Wg (wingless) [73]. Mutations in various components of Wnt lead to hyperactivation of Wnt signaling in HCC [74,75], which is ultimately accompanied by the release of β-catenin [76,77]. The activated β-catenin then translocates into the nucleus, where it triggers the transcription of several target genes [75], e.g., the LGR5 (leucine-rich repeat-containing G) receptor is associated with HCC metastasis [78,79]. It is known that about 40–70% of HCCs have β-catenin in the nucleus, increasing the signaling activity of Wnt/β-catenin [17,80,81,82]; however, its nuclear accumulation is limited to late-stage HCC, whereas in earlier stages, it is mainly localized to the plasma membrane in complex with several cadherin family members. This study highlights the important function of β-catenin during HCC progression, as it promotes tumor cell survival by enhancing signaling of growth factor receptors such as EGFR [83].
It has been shown that peptide-modified SeNPs (RGDfC-SeNPs) used to deliver siRNA (anti-Oct4) to liver cells can activate Wnt/β-catenin signaling. RGD peptide binds to αvβ3 integrin, which is overexpressed in cancer cells [84]. It was found that knockdown of Oct4 resulted in significant suppression of the proteins Sox-2 (a transcription factor required to maintain self-renewal or pluripotency of undifferentiated embryonic stem cells) and Nanog (a transcription factor involved in self-renewal of undifferentiated embryonic stem cells). It is known that the regulation of Oct4 expression is associated with the activation of Wnt/β-catenin signaling [85]. Oct4 knockdown led to a decrease in the expression of β -catenin and GSK-3β, an important regulator of β-catenin phosphorylation. These processes are shown schematically in Figure 2.
SeNPs can be an inducer of Wnt/β-catenin signaling activation in liver cancer cells. As an siRNA carrier, they cause knockdown of important transcription factor Oct4, which is accompanied by a decrease in the expression of transcription factors Sox-2 and Nanog, as well as a further decrease in the expression of β-catenin and GSK-3β, an important regulator of β-catenin phosphorylation; the end result of all these events is mitochondrially mediated apoptosis of liver cancer cells. In addition, SeNPs are able to reduce the expression of mTOR, AKT and PI3K, as well as activate LC3-II and p62, important participants in the processes occurring during autophagy, which leads to the death of cancer cells through autophagy.
It was also shown that SeNPs in combination with or without quercetin had anti-inflammatory and hepatoprotective effects in a rat model of thioacetamide-induced hepatocellular carcinoma. Suppression of HCC progression in rats was due to increased oxidative stress and dysregulation of the oncogenic p53/β-catenin/cyclin D signaling pathway [39].

4.3. Involvement of SeNPs in the Regulation of PI3K/Akt/mTOR in HCC

The PI3K/AKT/mTOR signaling pathway is associated with autophagy and apoptosis and plays a vital role in both processes [86,87,88,89]. Some anticancer drugs induce apoptosis and autophagy by inhibiting the PI3K/Akt/mTOR pathway [90,91,92]. There is also evidence of an interaction between autophagy and apoptosis through the Bcl-2 family and the PI3K/Akt/mTOR signaling pathway [93]. To date, there is sufficient evidence that various SeNPs are able to inhibit the PI3K/Akt/mTOR signaling pathway in various cancer cells [45,94,95,96]. These processes are shown schematically in Figure 2.
Chemically synthesized SeNPs complexed with curcumin (Se@Cur) contributed to a significant enhancement of the Akt protein, inducing apoptosis in HepG2 cells and activating the PI3K/Akt/mTOR pathway [27]. At the same time, after 24 h and especially after 48 h, SeNPs doped with sorafenib (SeSo) are able to reduce the expression of mTOR, AKT and PI3K [45]. It is known that mammalian rapamycin kinase mTOR is a negative central regulator of the autophagic process; therefore, inhibition of the PI3K/AKT/mTOR pathway leads to the activation of autophagy, which is also important evidence of the death of HepG2 cells through autophagy after 24 and 48 h of treatment with SeNPs and SeSo nanoparticles.

4.4. SeNPs Regulate the Expression of ER-Resident Selenoproteins in HCC

Recently, enough work has been accumulated demonstrating the important role of various selenium-containing compounds in the regulation of selenoprotein expression in various cancer cells [25,97,98,99,100,101,102,103,104,105,106,107,108]. Thus, we previously studied the patterns of expression of seven ER-resident selenoproteins by SeNPs doped (SeSo) and undoped (SeNPs) with sorafenib in HepG2 cells [45]. After 24 h of treating cells with SeNPs, an almost twofold decrease in the mRNA of selenoprotein SELENOM was observed, and a trend towards a decrease in mRNA expression was recorded for two other selenoproteins, namely SELENOS and SELENOF. The SeSo nanocomplex promoted an almost twofold increase in the expression of SELENOT mRNA and significant increases in SELENON and DIO2.
To date, enough data have been accumulated to allow us to say with confidence that SeNPs actively regulate the expression of these oxidoreductases. Thus, in a study of the chemoprotective properties of SeNPs during hepatotoxicity in mice induced by cyclophosphamide, they were found to contribute to an increase in the levels of glutathione peroxidases, along with those of other enzymes [38]. Similar data were obtained in the treatment of HCC with SeNPs and SeNPs doped with quercetin [109]. Our results should probably be considered a protective antioxidant response, which turns out to be insufficient under conditions of prolonged ER stress and severe inhibition of key kinase pathways and ultimately leads to apoptotic cell death to varying degrees, with a predominance of apoptosis over necrosis in the case of incubation of cells with SeNPs and SeSo [45].
There is evidence that SeNPs can prevent HCC and protect liver cells from disturbances in glutathione homeostasis; they were found to reverse the decrease in the activity of glutathione peroxidases and glutathione S-transferases in the liver but had a subtle effect on the activity of glutathione reductases [13].
Chitosan-stabilized SeNPs (CS-SeNPs) also demonstrated acceptable antioxidant activity, increasing the concentration of glutathione peroxidases in the liver and the level of reduced glutathione in the serum of mice after injections with concavalin A, which indicates the hepatoprotective function of Cs-SeNPs, including protection against HCC [12].

5. Use of SeNPs to Improve the Effectiveness of Drugs in the Treatment of HCC

Radiation therapy and chemotherapy are both traditional methods of treating HCC. However, a number of problems are associated with multidrug resistance, targeted delivery and effective release of drugs, which makes these treatments highly ineffective.
Lipiodol chemoembolization (LCE) is widely used to treat patients with unresectable HCC [110], but it has poor control over sustained drug release [111,112]. In addition, So is a first-line systemic drug that significantly increases the survival of patients with HCC [113,114]. It inhibits tumor angiogenesis by blocking VEGF and PDGF receptors and tumor cell proliferation by blocking the B-Raf and c-Raf MAP kinase pathways to accelerate tumor cell apoptosis [115,116,117]. However, this drug has a number of serious side effects due to nonspecific absorption by normal tissues [118,119]. To improve anticancer effectiveness, an injectable nanosystem based on a thermosensitive hydrogel containing SeNPs and So was developed [120]. At the cellular level, it was shown that this hydrogel reduced the expression of CD34 and Ki67 and increased the growth of caspase-3 in tumor tissue. In vivo experiments showed that the developed thermosensitive nanosystem had great potential for local application, in addition to non-obvious side effects and low toxicity for nude mice. Treating mice with radiation therapy and the hydrogel together resulted in improvements in protein, serum creatinine, cholesterol and blood glucose levels, and no abnormalities in major organ function were observed in the mice.
In another study, in vivo experiments found that sorafenib-doped SeNPs restored the effects of TAA injections in animals; they reduced oxidative stress and caused the activation of pro-apoptotic genes encoding p53, BAX and caspase-3, as well as the inhibition of the expression of anti-apoptotic gene Bcl-2. Nanoselenium enhanced the effectiveness of sorafenib and reduced the resistance of liver cancer cells to drugs, affecting the mTOR and NF-kB pathways, in addition to contributing to a decrease in angiogenesis and metastasis [121].
Another common and effective chemotherapeutic drug, including for the treatment of HCC, is doxorubicin, but its clinical use is limited due to poor water solubility and off-target side effects [122,123]. To effectively deliver doxorubicin to liver malignancy, galactose-modified SeNPs loaded with doxorubicin were developed and demonstrated excellent cellular uptake into HepG2 cells via clathrin-mediated endocytosis, accompanied by rapid release of doxorubicin from the nanoparticles at pH 5.4. This nanocomplex more effectively suppressed HepG2 cell proliferation and induced apoptosis through activation of caspase-3 pathways. In vivo studies showed that the resulting nanocomplex reduced the percentage of Ki67-positive cancer cells. Since nuclear protein Ki67 is associated with cell proliferation, the nanocomplex reduced the proliferation of cancer cells and the size of tumors in mice. In addition, no pathological changes were observed in the heart, liver, spleen, lungs or kidneys of mice after treatment with the studied nanocomplex, which indicates the absence of its toxicity in vivo [124].
The main molecular mechanisms activated by various SeNPs in in vitro and in vivo HCC models are presented in Table 1.

6. Conclusions

This review presents currently available data on the molecular mechanisms regulating processes associated with the cytotoxic effects of SeNPs of various origins in hepatocellular carcinoma. The signaling cascades that are activated after exposure of liver cancer cells to selenium nanoparticles and the role of selenoproteins in these processes are described, the available biomarkers of HCC are described and the concept of intratumoral heterogeneity in HCC is considered. This review allows us to understand the selectivity of SeNPs against HCC, which is extremely important for the development of targeted hepatoprotective drugs based on them.

Funding

This study was supported by the Russian Science Foundation (grant No. 23-25-00030; https://rscf.ru/project/23-25-00030/). Grant was accessed on 13 January 2023.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Nabil, A.-A.; Charles, S.; Alberto, B. Tumor Evolution as a Therapeutic Target. Cancer Discov. 2017, 7, 805–817. [Google Scholar]
  2. Kurebayashi, Y.; Ojima, H.; Tsujikawa, H.; Kubota, N.; Maehara, J.; Abe, Y.; Kitago, M.; Shinoda, M.; Kitagawa, Y.; Sakamoto, M. Landscape of immune microenvironment in hepatocellular carcinoma and its additional impact on histological and molecular classification. Hepatology 2018, 68, 1025–1041. [Google Scholar] [CrossRef]
  3. Losic, B.; Craig, A.J.; Villacorta-Martin, C.; Martins-Filho, S.N.; Akers, N.; Chen, X.; Ahsen, M.E.; von Felden, J.; Labgaa, I.; DʹAvola, D.; et al. Intratumoral heterogeneity and clonal evolution in liver cancer. Nat. Commun. 2020, 11, 291. [Google Scholar] [CrossRef]
  4. Guo, L.; Yi, X.; Chen, L.; Zhang, T.; Guo, H.; Chen, Z.; Cheng, J.; Cao, Q.; Liu, H.; Hou, C.; et al. Single-Cell DNA Sequencing Reveals Punctuated and Gradual Clonal Evolution in Hepatocellular Carcinoma. Gastroenterology 2022, 162, 238–252. [Google Scholar] [CrossRef] [PubMed]
  5. Bruix, J.; Qin, S.; Merle, P.; Granito, A.; Huang, Y.H.; Bodoky, G.; Pracht, M.; Yokosuka, O.; Rosmorduc, O.; Breder, V.; et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 389, 56–66. [Google Scholar] [CrossRef]
  6. Abou-Alfa, G.K.; Meyer, T.; Cheng, A.L.; El-Khoueiry, A.B.; Rimassa, L.; Ryoo, B.Y.; Cicin, I.; Merle, P.; Chen, Y.; Park, J.W.; et al. Cabozantinib in patients with advanced progressing hepatocellular carcinoma. N. Engl. J. Med. 2018, 379, 54–63. [Google Scholar] [CrossRef]
  7. Zhu, A.X.; Kang, Y.K.; Yen, C.J.; Finn, R.S.; Galle, P.R.; Llovet, J.M.; Assenat, E.; Brandi, G.; Pracht, M.; Lim, H.Y.; et al. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased alpha-fetoprotein concentrations (REACH-2): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019, 20, 282–296. [Google Scholar] [CrossRef] [PubMed]
  8. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef]
  9. Mintz, K.; Waidely, E.; Zhou, Y.; Peng, Z.; Al-Youbi, A.O.; Bashammakh, A.S.; El-Shahawi, M.S.; Leblanc, R.M. Carbon dots and gold nanoparticles based immunoassay for detection of alpha-L-fucosidase. Anal. Chim. Acta. 2018, 1041, 114–121. [Google Scholar] [CrossRef] [PubMed]
  10. Ma, X.-H.; Wang, S.; Liu, S.-Y.; Chen, K.; Wu, Z.-Y.; Li, D.-F.; Mi, Y.-T.; Hu, L.-B.; Chen, Z.-W.; Zhao, X.-M. Development and in vitro study of a bi-specific magnetic resonance imaging molecular probe for hepatocellular carcinoma. World J. Gastroenterol. 2019, 25, 3030–3043. [Google Scholar] [CrossRef]
  11. Mohammed, E.; El-Beih, N.; El-Hussieny, E.; El-Ahwany, E.; Hassan, M.; Zoheiry, M. Effects of free and nanoparticulate curcumin on chemically induced liver carcinoma in an animal model. Arch. Med. Sci. 2021, 17, 218–227. [Google Scholar] [CrossRef]
  12. Bai, K.; Hong, B.; He, J.; Huang, W. Antioxidant Capacity and Hepatoprotective Role of Chitosan-Stabilized SeNPs in Concanavalin A-Induced Liver Injury in Mice. Nutrients 2020, 12, 857. [Google Scholar] [CrossRef]
  13. Qiu, Y.; Chen, X.; Chen, Z.; Zeng, X.; Yue, T.; Yuan, Y. Effects of SeNPs on Preventing Patulin-Induced Liver, Kidney and Gastrointestinal Damage. Foods 2022, 11, 749. [Google Scholar] [CrossRef]
  14. Hamza, R.Z.; Al-Motaan, S.E.; Malik, N. Protective and Antioxidant Role of SeNPs and Vitamin C against Acrylamide Induced Hepatotoxicity in Male Mice. Int. J. Pharmacol. 2019, 15, 664–674. [Google Scholar] [CrossRef]
  15. Bhattacharjee, A.; Basu, A.; Ghosh, P.; Biswas, J.; Bhattacharya, S. Protective effect of Selenium nanoparticle against cyclophosphamide induced hepatotoxicity and genotoxicity in Swiss albino mice. J. Biomater. Appl. 2014, 29, 303–317. [Google Scholar] [CrossRef]
  16. Khan, M.A.; Singh, D.; Arif, A.; Sodhi, K.K.; Singh, D.K.; Islam, S.N.; Ahmad, A.; Akhtar, K.; Siddique, H.R. Protective effect of green synthesized SeNPs against Doxorubicin induced multiple adverse effects in Swiss albino mice. Life Sci. 2022, 305, 120792. [Google Scholar] [CrossRef]
  17. Nhieu, J.T.; Renard, C.A.; Wei, Y.; Cherqui, D.; Zafrani, E.S.; Buendia, M.A. Nuclear accumulation of mutated beta-catenin in hepatocellular carcinoma is associated with increased cell proliferation. Am. J. Pathol. 1999, 155, 703–710. [Google Scholar] [CrossRef]
  18. Salehi, B.; Martorell, M.; Arbiser, J.L.; Sureda, A.; Martins, N.; Maurya, P.K.; Sharifi-Rad, M.; Kumar, P.; Sharifi-Rad, J. Antioxidants: Positive or Negative Actors? Biomolecules 2018, 8, 124. [Google Scholar] [CrossRef]
  19. MacFarquhar, J.K.; Broussard, D.L.; Melstrom, P.; Hutchinson, R.; Wolkin, A.; Martin, C.; Burk, R.F.; Dunn, J.R.; Green, A.L.; Hammond, R.; et al. Acute selenium toxicity associated with a dietary supplement. Arch. Intern. Med. 2010, 170, 256–261. [Google Scholar] [CrossRef]
  20. Xiao, X.; Deng, H.; Lin, X.; Ali, A.S.M.; Viscardi, A.; Guo, Z.; Qiao, L.; He, Y.; Han, J. Selenium nanoparticles: Properties, preparation methods, and therapeutic applications. Chem. Biol. Interact. 2023, 378, 110483. [Google Scholar] [CrossRef]
  21. Chen, N.; Yao, P.; Zhang, W.; Zhang, Y.; Xin, N.; Wei, H.; Zhang, T.; Zhao, C. Selenium nanoparticles: Enhanced nutrition and beyond. Crit. Rev. Food Sci. Nutr. 2023, 63, 12360–12371. [Google Scholar] [CrossRef]
  22. Varlamova, E.G.; Gudkov, S.V.; Plotnikov, E.Y.; Turovsky, E.A. Size-Dependent Cytoprotective Effects of Selenium Nanoparticles during Oxygen-Glucose Deprivation in Brain Cortical Cells. Int. J. Mol. Sci. 2022, 23, 7464. [Google Scholar] [CrossRef]
  23. Zhai, X.; Zhang, C.; Zhao, G.; Stoll, S.; Ren, F.; Leng, X. Antioxidant capacities of the selenium nanoparticles stabilized by chitosan. J. Nanobiotechnology 2017, 15, 4. [Google Scholar] [CrossRef]
  24. Menon, S.; Ks, S.D.; R, S.; S, R.; S, V.K. Selenium nanoparticles: A potent chemotherapeutic agent and an elucidation of its mechanism. Colloids Surf. B Biointerfaces 2018, 170, 280–292. [Google Scholar] [CrossRef]
  25. Varlamova, E.G.; Goltyaev, M.V.; Mal’tseva, V.N.; Turovsky, E.A.; Sarimov, R.M.; Simakin, A.V.; Gudkov, S.V. Mechanisms of the Cytotoxic Effect of Selenium Nanoparticles in Different Human Cancer Cell Lines. Int. J. Mol. Sci. 2021, 22, 7798. [Google Scholar] [CrossRef]
  26. Huang, Y.; He, L.; Liu, W.; Fan, C.; Zheng, W.; Wong, Y.S.; Chen, T. Selective cellular uptake and induction of apoptosis of cancer-targeted selenium nanoparticles. Biomaterials 2013, 34, 7106–7116. [Google Scholar] [CrossRef]
  27. Guo, M.; Li, Y.; Lin, Z.; Zhao, M.; Xiao, M.; Wang, C.; Xu, T.; Xiaa, Y.; Zhu, B. Surface decoration of selenium nanoparticles with curcumin induced HepG2 cell apoptosis through ROS mediated p53 and AKT signaling pathways. RSC Adv. 2017, 7, 52456–52464. [Google Scholar] [CrossRef]
  28. Sonkusre, P.; Cameotra, S.S. Biogenic selenium nanoparticles induce ROS-mediated necroptosis in PC-3 cancer cells through TNF activation. J. Nanobiotechnol. 2017, 15, 43. [Google Scholar] [CrossRef]
  29. Pi, J.; Jiang, J.; Cai, H.; Yang, F.; Jin, H.; Yang, P.; Cai, J.; Chen, Z.W. GE11 peptide conjugated selenium nanoparticles for EGFR targeted oridonin delivery to achieve enhanced anticancer efficacy by inhibiting EGFR-mediated PI3K/AKT and Ras/Raf/MEK/ERK pathways. Drug Deliv. 2017, 24, 1549–1564. [Google Scholar] [CrossRef]
  30. Deng, X.; Liu, H.; Xu, Y.; Chan, L.; Xie, J.; Xiong, Z.; Tang, Z.; Yang, F.; Chen, T. Designing highly stable ferrous selenide-black phosphorus nanosheets heteronanostructure via p-Se bond for MRI-guided photothermal therapy. J. Nanobiotechnol. 2021, 19, 201. [Google Scholar] [CrossRef]
  31. Liu, W.; Su, J.; Shi, Q.; Wang, J.; Chen, X.; Zhang, S.; Li, M.; Cui, J.; Fan, C.; Sun, B.; et al. RGD peptide-conjugated selenium nanocomposite inhibits human glioma growth by triggering mitochondrial dysfunction and ROS-dependent MAPKs activation. Front. Bioeng. Biotechnol. 2021, 9, 781608. [Google Scholar] [CrossRef]
  32. Mi, X.J.; Choi, H.S.; Perumalsamy, H.; Shanmugam, R.; Thangavelu, L.; Balusamy, S.R.; Kim, Y.J. Biosynthesis and cytotoxic effect of silymarin-functionalized selenium nanoparticles induced autophagy mediated cellular apoptosis via downregulation of PI3K/Akt/mTOR pathway in gastric cancer. Phytomedicine 2022, 99, 154014. [Google Scholar] [CrossRef]
  33. Bjorkoy, G.; Lamark, T.; Pankiv, S.; Overvatn, A.; Brech, A.; Johansen, T. Monitoring autophagic degradation of p62/SQSTM1. Methods Enzymol. 2009, 452, 181–197. [Google Scholar] [CrossRef]
  34. Huang, G.; Liu, Z.; He, L.; Luk, K.H.; Cheung, S.T.; Wong, K.H.; Chen, T. Autophagy is an important action mode for functionalized selenium nanoparticles to exhibit anti-colorectal cancer activity. Biomater. Sci. 2018, 6, 2508–2517. [Google Scholar] [CrossRef]
  35. Huang, J.; Liu, Y.; Liu, T.; Chang, Y.; Chen, T.; Li, X. Dual-targeting nanotherapeutics antagonize hyperinsulinemia-promoted tumor growth via activating cell autophagy. J. Mater. Chem. B 2019, 7, 6751–6758. [Google Scholar] [CrossRef]
  36. Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466. [Google Scholar] [CrossRef]
  37. 37. Cui, D.; Ma, J.; Liang, T.; Sun, L.; Meng, L.; Liang, T.; Li, Q. Selenium nanoparticles fabricated in laminarin polysaccharides solutions exert their cytotoxicities in HepG2 cells by inhibiting autophagy and promoting apoptosis. Int. J. Biol. Macromol. 2019, 137, 829–835. [Google Scholar] [CrossRef]
  38. Mohamed, A.A.; Zaghloul, R.A.; Abdelghany, A.M.; El Gayar, A.M. Selenium nanoparticles and quercetin suppress thioacetamide-induced hepatocellular carcinoma in rats: Attenuation of inflammation involvement. J. Biochem. Mol. Toxicol. 2022, 36, e22989. [Google Scholar] [CrossRef]
  39. Hadrup, N.; Ravn-Haren, G. Toxicity of repeated oral intake of organic selenium, inorganic selenium, and selenium nanoparticles: A review. J. Trace Elem. Med. Biol. 2023, 79, 127235. [Google Scholar] [CrossRef]
  40. Jia, X.; Li, N.; Chen, J. A subchronic toxicity study of elemental Nano-Se in Sprague-Dawley rats. Life Sci. 2005, 76, 1989–2003. [Google Scholar] [CrossRef]
  41. Lesnichaya, M.; Karpova, E.; Sukhov, B. Effect of high dose of selenium nanoparticles on antioxidant system and biochemical profile of rats in correction of carbon tetrachloride-induced toxic damage of liver. Colloids Surf. B Biointerfaces 2021, 197, 111381. [Google Scholar] [CrossRef]
  42. Ji, H.; Lou, X.; Jiao, J.; Li, Y.; Dai, K.; Jia, X. Preliminary Structural Characterization of Selenium Nanoparticle Composites Modified by Astragalus Polysaccharide and the Cytotoxicity Mechanism on Liver Cancer Cells. Molecules 2023, 28, 1561. [Google Scholar] [CrossRef]
  43. Zheng, S.; Li, X.; Zhang, Y.; Xie, Q.; Wong, Y.S.; Zheng, W.; Chen, T. PEG-nanolized ultrasmall SeNPs overcome drug resistance in hepatocellular carcinoma HepG2 cells through induction of mitochondria dysfunction. Int. J. Nanomed. 2012, 7, 3939–3949. [Google Scholar]
  44. Jiao, J.; Yu, J.; Ji, H.; Liu, A. Synthesis of macromolecular Astragalus polysaccharide-nano selenium complex and the inhibitory effects on HepG2 cells. Int. J. Biol. Macromol. 2022, 211, 481–489. [Google Scholar] [CrossRef]
  45. Varlamova, E.G.; Goltyaev, M.V.; Simakin, A.V.; Gudkov, S.V.; Turovsky, E.A. Comparative Analysis of the Cytotoxic Effect of a Complex of SeNPs Doped with Sorafenib, “Naked” Selenium Nanoparticles, and Sorafenib on Human Hepatocyte Carcinoma HepG2 Cells. Int. J. Mol. Sci. 2022, 23, 6641. [Google Scholar] [CrossRef]
  46. Li, Y.; Guo, M.; Lin, Z.; Zhao, M.; Xia, Y.; Wang, C.; Xu, T.; Zhu, B. Multifunctional SeNPs with Galangin-induced HepG2 cell apoptosis through p38 and AKT signalling pathway. R. Soc. Open Sci. 2018, 5, 180509. [Google Scholar] [CrossRef]
  47. Xia, Y.; Guo, M.; Xu, T.; Li, Y.; Wang, C.; Lin, Z.; Zhao, M.; Zhu, B. siRNA-loaded selenium nanoparticle modified with hyaluronic acid for enhanced hepatocellular carcinoma therapy. Int. J. Nanomed. 2018, 13, 1539–1552. [Google Scholar] [CrossRef] [PubMed]
  48. Li, Y.; Lin, Z.; Zhao, M.; Xu, T.; Wang, C.; Xia, H.; Wang, H.; Zhu, B. Multifunctional SeNPs as carriers of HSP70 siRNA to induce apoptosis of HepG2 cells. Int. J. Nanomed. 2016, 11, 3065–3076. [Google Scholar]
  49. Cui, D.; Liang, T.; Sun, L.; Meng, L.; Yang, C.; Wang, L.; Liang, T.; Li, Q. Green synthesis of SeNPs with extract of hawthorn fruit induced HepG2 cells apoptosis. Pharm. Biol. 2018, 56, 528–534. [Google Scholar] [CrossRef] [PubMed]
  50. He, Z.; Song, H.; Cheng, J.; Qiu, W.; Hu, J.; Qiu, Z.; Wang, Q.; Chang, C.; Zheng, G.; Meng, Y. SeNPs Stabilized by β-Glucan Nanotubes from Black Fungus and Their Effects on the Proliferation, Apoptosis, and Cell Cycle of HepG2 Cells. ACS Omega 2023, 8, 45358–45368. [Google Scholar] [CrossRef] [PubMed]
  51. Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef]
  52. Yang, Z.; Klionsky, D.J. Eaten alive: A history of macroautophagy. Nat. Cell Biol. 2010, 12, 814–822. [Google Scholar] [CrossRef]
  53. Pankiv, S.; Clausen, T.H.; Lamark, T.; Brech, A.; Bruun, J.A.; Outzen, H.; Overvatn, A.; Bjorkoy, G.; Johansen, T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 2007, 282, 24131–24145. [Google Scholar] [CrossRef] [PubMed]
  54. Bjorkoy, G.; Lamark, T.; Brech, A.; Outzen, H.; Perander, M.; Overvatn, A.; Stenmark, H.; Johansen, T. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J. Cell Biol. 2005, 171, 603–614. [Google Scholar] [CrossRef] [PubMed]
  55. Tsai, Y.; Weissman, A. The Unfolded Protein Response, Degradation from the Endoplasmic Reticulum, and Cancer. Genes. Cancer 2010, 1, 764–778. [Google Scholar] [CrossRef]
  56. Marciniak, S.; Ron, D. Endoplasmic Reticulum Stress Signaling in Disease. Physiol. Rev. 2006, 86, 1133–1149. [Google Scholar] [CrossRef]
  57. Harding, H.P.; Zhang, Y.; Zeng, H.; Novoa, I.; Lu, P.D.; Calfon, M. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol. Cell 2003, 11, 619–633. [Google Scholar] [CrossRef]
  58. Lu, P.D.; Harding, H.P.; Ron, D. Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response. J. Cell Biol. 2004, 167, 27–33. [Google Scholar] [CrossRef] [PubMed]
  59. Rutkowski, D.T.; Kaufman, R.J. All roads lead to ATF4. Dev. Cell 2003, 4, 442–444. [Google Scholar] [CrossRef]
  60. Vattem, K.M.; Wek, R.C. Reinitiation involving upstream ORFs regulates ATF-4 mRNA translation in mammalian cells. Proc. Natl. Acad. Sci. USA 2004, 101, 11269–11274. [Google Scholar] [CrossRef]
  61. Calfon, M.; Zeng, H.; Urano, F.; Till, J.H.; Hubbard, S.R.; Harding, H.P.; Clark, S.G.; Ron, D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 2002, 415, 92–96. [Google Scholar] [CrossRef] [PubMed]
  62. Yoshida, H.; Matsui, T.; Yamamoto, A.; Okada, T.; Mori, K. XBP1 mRNA is induced by ATF-6 and spliced by IRE1 in response to ER-stress to produce a highly active transcription factor. Cell 2001, 107, 881–891. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, A.H.; Chu, G.C.; Iwakoshi, N.N.; Glimcher, L.H. XBP-1 is required for biogenesis of cellular secretory machinery of exocrine glands. EMBO J. 2005, 24, 4368–4380. [Google Scholar] [CrossRef] [PubMed]
  64. Orrenius, S.; Zhivotovsky, B.; Nicotera, P. Regulation of cell death: The calcium-apoptosis link. Nat. Rev. Mol. Cell. Biol. 2003, 4, 552–565. [Google Scholar] [CrossRef] [PubMed]
  65. Ryan, P.M.; Bedard, K.; Breining, T.; Cribb, A.E. Disruption of the endoplasmic reticulum by cytotoxins in LLC-PK1 cells. Toxicol. Lett. 2005, 159, 154–163. [Google Scholar] [CrossRef] [PubMed]
  66. Tan, Y.; Dourdin, N.; Wu, C.; De Veyra, T.; Elce, J.S.; Greer, P.A. Ubiquitous calpains promote caspase-12 and JNK activation during endoplasmic reticulum stress-induced apoptosis. J. Biol. Chem. 2006, 281, 16016–16024. [Google Scholar] [CrossRef] [PubMed]
  67. Scorrano, L.; Oakes, S.A.; Opferman, J.T.; Cheng, E.H.; Sorcinelli, M.D.; Pozzan, T.; Korsmeyer, S.J. BAX and BAK regulation of endoplasmic reticulum Ca2+: A control point for apoptosis. Science 2003, 300, 135–139. [Google Scholar] [CrossRef] [PubMed]
  68. Zong, W.X.; Li, C.; Hatzivassiliou, G.; Lindsten, T.; Yu, Q.C.; Yuan, J.; Thompson, C.B. Bax and Bak can localize to the endoplasmic reticulum to initiate apoptosis. J. Cell Biol. 2003, 162, 59–69. [Google Scholar] [CrossRef] [PubMed]
  69. Hitomi, J.; Katayama, T.; Eguchi, Y.; Kudo, T.; Taniguchi, M.; Koyama, Y.; Manabe, T.; Yamagishi, S.; Bando, Y.; Imaizumi, K.; et al. Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Abeta-induced cell death. J. Cell Biol. 2004, 165, 347–356. [Google Scholar] [CrossRef]
  70. Schroder, M.; Kaufman, R.J. The mammalian unfolded protein response. Ann. Rev. Biochem. 2005, 74, 739–789. [Google Scholar] [CrossRef]
  71. Anding, A.L.; Chapman, J.S.; Barnett, D.W.; Curley RWJr Clagett-Dame, M. The unhydrolyzable fenretinide analogue 4-hydroxybenzylretinone induces the proapoptotic genes GADD153 (CHOP) and Bcl-2-binding component 3 (PUMA) and apoptosis that is caspase- dependent and independent of the retinoic acid receptor. Cancer Res. 2007, 67, 6270–6277. [Google Scholar] [CrossRef] [PubMed]
  72. McCullough, K.D.; Martindale, J.L.; Klotz, L.O.; Aw, T.Y.; Holbrook, N.J. Gadd 153 sensitizes cells by downregulating Bcl2 and perturbing the cellular redox state. Mol. Cell Biol. 2001, 21, 1249–1259. [Google Scholar] [CrossRef] [PubMed]
  73. Yang, Y.; Ye, Y.-C.; Chen, Y.; Zhao, J.-L.; Gao, C.-C.; Han, H.; Liu, W.-C.; Qin, H.-Y. Crosstalk between hepatic tumor cells and macrophages via Wnt/β-catenin signaling promotes M2-like macrophage polarization and reinforces tumor malignant behaviors. Cell Death Dis. 2018, 9, 793. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, W.; Smits, R.; Hao, H.; He, C. Wnt/β-Catenin Signaling in Liver Cancers. Cancers 2019, 11, 926. [Google Scholar] [CrossRef] [PubMed]
  75. Xu, W.; Zhou, W.; Cheng, M.; Wang, J.; Liu, Z.; He, S.; Luo, X.; Huang, W.; Chen, T.; Yan, W.; et al. Hypoxia activates Wnt/β-catenin signaling by regulating the expression of BCL9 in human hepatocellular carcinoma. Sci. Rep. 2017, 7, 40446. [Google Scholar] [CrossRef] [PubMed]
  76. Stamos, J.L.; Weis, W.I. The β-catenin destruction complex. Cold Spring Harb. Perspect. Biol. 2013, 5, a007898. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, C.; Takada, K.; Zhu, D. Targeting Wnt/β-catenin pathway for drug therapy. Med. Drug Discov. 2020, 8, 100066. [Google Scholar] [CrossRef]
  78. Ma, Z.; Guo, D.; Wang, Q.; Liu, P.; Xiao, Y.; Wu, P.; Wang, Y.; Chen, B.; Liu, Z.; Liu, Q. Lgr5-mediated p53 Repression through PDCD5 leads to doxorubicin resistance in Hepatocellular Carcinoma. Theranostics 2019, 9, 2967. [Google Scholar] [CrossRef] [PubMed]
  79. Koni, M.; Pinnarò, V.; Brizzi, M.F. The Wnt signalling pathway: A tailored target in cancer. Int. J. Mol. Sci. 2020, 21, 7697. [Google Scholar] [CrossRef]
  80. Lachenmayer, A.; Alsinet, C.; Savic, R.; Cabellos, L.; Toffanin, S.; Hoshida, Y.; Villanueva, A.; Minguez, B.; Newell, P.; Tsai, H.W.; et al. Wnt-pathway activation in two molecular classes of hepatocellular carcinoma and experimental modulation by sorafenib. Clin. Cancer Res. 2012, 18, 4997–5007. [Google Scholar] [CrossRef]
  81. Wong, C.M.; Fan, S.T.; Ng, I.O. beta-Catenin mutation and overexpression in hepatocellular carcinoma: Clinicopathologic and prognostic significance. Cancer 2001, 92, 136–145. [Google Scholar] [CrossRef] [PubMed]
  82. Lin, Y.T.; Chao, C.C. Identification of the beta-catenin/JNK/prothymosin-alpha axis as a novel target of sorafenib in hepatocellular carcinoma cells. Oncotarget 2015, 6, 38999–39017. [Google Scholar] [CrossRef] [PubMed]
  83. Kim, E.; Lisby, A.; Ma, C.; Lo, N.; Ehmer, U.; Hayer, K.E.; Furth, E.E.; Viatour, P. Promotion of growth factor signaling as a critical function of β-catenin during HCC progression. Nat. Commun. 2019, 10, 1909. [Google Scholar] [CrossRef] [PubMed]
  84. Xia, Y.; Lin, Z.; Li, Y.; Zhao, M.; Wang, C.; Guo, M.; Zhang, B.; Zhu, B. Targeted delivery of siRNA using RGDfC-conjugated functionalized SeNPs for anticancer therapy. J. Mater. Chem. B 2017, 5, 6941–6952. [Google Scholar] [CrossRef] [PubMed]
  85. Zhu, J.Y.; Yang, X.; Chen, Y.; Jiang, Y.; Wang, S.J.; Li, Y.; Wang, X.Q.; Meng, Y.; Zhu, M.M.; Ma, X.; et al. Phytother. Res. 2017, 31, 680–688. [Google Scholar]
  86. Alzahrani, A.S. PI3K/Akt/mTOR inhibitors in cancer: At the bench and bedside. Semin. Cancer Biol. 2019, 59, 125–132. [Google Scholar] [CrossRef] [PubMed]
  87. Pompura, S.L.; Dominguez-Villar, M. The PI3K/AKT signaling pathway in regulatory T-cell development, stability, and function. J. Leukocyte Biol. 2018, 103, 1065–1076. [Google Scholar] [CrossRef] [PubMed]
  88. Li, H.; Prever, L.; Hirsch, E.; Gulluni, F. Targeting PI3K/AKT/mTOR signaling pathway in breast cancer. Cancers 2021, 13, 3517. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, L.; Wang, H.; Xu, J.; Zhu, J.; Ding, K. Inhibition of cathepsin S induces autophagy and apoptosis in human glioblastoma cell lines through ROS-mediated PI3K/AKT/mTOR/p70S6K and JNK signaling pathways. Toxicol. Lett. 2014, 228, 248–259. [Google Scholar] [CrossRef]
  90. Wang, G.; Zhang, T.; Sun, W.; Wang, H.; Yin, F.; Wang, Z.; Zuo, D.; Sun, M.; Zhou, Z.; Lin, B.; et al. Arsenic sulfide induces apoptosis and autophagy through the activation of ROS/JNK and suppression of Akt/mTOR signaling pathways in osteosarcoma. Free Radic. Biol. Med. 2017, 106, 24–37. [Google Scholar] [CrossRef]
  91. Ma, K.; Zhang, C.; Huang, M.Y.; Li, W.Y.; Hu, G.Q. Cinobufagin induces autophagy-mediated cell death in human osteosarcoma U2OS cells through the ROS/JNK/p38 signaling pathway. Oncol. Rep. 2016, 36, 90–98. [Google Scholar] [CrossRef] [PubMed]
  92. Kim, S.H.; Son, K.M.; Kim, K.Y.; Yu, S.N.; Park, S.G.; Kim, Y.W.; Nam, H.W.; Suh, J.T.; Ji, J.H.; Ahn, S.C. Deoxypodophyllotoxin induces cytoprotective autophagy against apoptosis via inhibition of PI3K/AKT/mTOR pathway in osteosarcoma U2OS cells. Pharmacol. Rep. 2017, 69, 878–884. [Google Scholar] [CrossRef] [PubMed]
  93. Pan, S.T.; Qin, Y.; Zhou, Z.W.; He, Z.X.; Zhang, X.; Yang, T.; Yang, Y.X.; Wang, D.; Qiu, J.X.; Zhou, S.F. Plumbagin induces G2/M arrest, apoptosis, and autophagy via p38 MAPK-and PI3K/Akt/mTOR-mediated pathways in human tongue squamous cell carcinoma cells. Drug Des. Dev. Ther. 2015, 9, 1601. [Google Scholar]
  94. Wang, R.; Ha, K.Y.; Dhandapani, S.; Kim, Y.J. Biologically synthesized black ginger-selenium nanoparticle induces apoptosis and autophagy of AGS gastric cancer cells by suppressing the PI3K/Akt/mTOR signaling pathway. J. Nanobiotechnology 2022, 20, 441. [Google Scholar] [CrossRef] [PubMed]
  95. Mi, X.J.; Le, H.M.; Lee, S.; Park, H.R.; Kim, Y.J. Silymarin-Functionalized SeNPs Prevent LPS-Induced Inflammatory Response in RAW264.7 Cells through Downregulation of the PI3K/Akt/NF-κB Pathway. ACS Omega 2022, 7, 42723–42732. [Google Scholar] [CrossRef] [PubMed]
  96. Rabah, H.M.; Mohamed, D.A.; Mariah, R.A.; Abd El-Khalik, S.R.; Khattab, H.A.; AbuoHashish, N.A.; Abdelsattar, A.M.; Raslan, M.A.; Farghal, E.E.; Eltokhy, A.K. Novel insights into the synergistic effects of SeNPs and metformin treatment of letrozole—Induced polycystic ovarian syndrome: Targeting PI3K/Akt signalling pathway, redox status and mitochondrial dysfunction in ovarian tissue. Redox Rep. 2023, 28, 2160569. [Google Scholar] [CrossRef] [PubMed]
  97. Kuznetsova, Y.P.; Goltyaev, M.V.; Gorbacheva, O.S.; Novoselov, S.V.; Varlamova, E.G.; Fesenko, E.E. Influence of Sodium Selenite on the mRNA Expression of the Mammalian Selenocysteine-Containing Protein Genes in Testicle and Prostate Cancer Cells. Dokl. Biochem. Biophys. 2018, 480, 131–134. [Google Scholar] [CrossRef] [PubMed]
  98. Varlamova, E.G. Participation of selenoproteins localized in the ER in the processes occurring in this organelle and in the regulation of carcinogenesis-associated processes. J. Trace Elem. Med. Biol. 2018, 48, 172–180. [Google Scholar] [CrossRef] [PubMed]
  99. Varlamova, E.G.; Goltyaev, M.V.; Kuznetsova, J.P. Effect of Sodium Selenite on Gene Expression of SELF, SELW, and TGR Selenoproteins in Adenocarcinoma Cells of the Human Prostate. Mol. Biol. 2018, 52, 519–526. (In Russian) [Google Scholar] [CrossRef]
  100. Goltyaev, M.V.; Mal’tseva, V.N.; Varlamova, E.G. Expression of ER-resident selenoproteins and activation of cancer cells apoptosis mechanisms under ER-stress conditions caused by methylseleninic acid. Gene 2020, 755, 144884. [Google Scholar] [CrossRef]
  101. Goltyaev, M.V.; Varlamova, E.G.; Novoselov, S.V.; Fesenko, E.E. Activation of Signal Pathways of Apoptosis under Conditions of Prolonged ER-Stress Caused by Exposure of Mouse Testicular Teratoma Cells to Selenium-Containing Compounds. Dokl. Biochem. Biophys. 2020, 490, 9–11. [Google Scholar] [CrossRef] [PubMed]
  102. Varlamova, E.G.; Turovsky, E.A. The main cytotoxic effects of methylseleninic acid on various cancer cells. Int. J. Mol. Sci. 2021, 22, 6614. [Google Scholar] [CrossRef] [PubMed]
  103. Pitts, M.W.; Hoffmann, P.R. Endoplasmic reticulum–resident selenoproteins as regulators of calcium signaling and homeostasis. Cell Calcium. 2018, 70, 76–86. [Google Scholar] [CrossRef] [PubMed]
  104. Khurana, A.; Tekula, S.; Saifi, M.A.; Venkatesh, P.; Godugu, C. Therapeutic applications of selenium nanoparticles. Biomed. Pharmacother. 2019, 111, 802–812. [Google Scholar] [CrossRef] [PubMed]
  105. Turovsky, E.A.; Varlamova, E.G. Mechanism of Ca2+-Dependent Pro-Apoptotic Action of Selenium Nanoparticles, Mediated by Activation of Cx43 Hemichannels. Biology 2021, 10, 743. [Google Scholar] [CrossRef] [PubMed]
  106. Mal’tseva, V.N.; Goltyaev, M.V.; Turovsky, E.A.; Varlamova, E.G. Immunomodulatory and Anti-Inflammatory Properties of Selenium-Containing Agents: Their Role in the Regulation of Defense Mechanisms against COVID-19. Int. J. Mol. Sci. 2022, 23, 2360. [Google Scholar] [CrossRef]
  107. Mal’tseva, V.N.; Goltyaev, M.V.; Novoselov, S.V.; Varlamova, E.G. Effects of Sodium Selenite and Dithiothreitol on Expression of Endoplasmic Reticulum Selenoproteins and Apoptosis Markers in MSF7 Breast Adenocarcinoma Cells. Mol. Biol. 2022, 56, 135–146. (In Russian) [Google Scholar] [CrossRef]
  108. Varlamova, E.G.; Goltyaev, M.V.; Novoselov, V.I.; Fesenko, E.E. Cloning, intracellular localization, and expression of the mammalian selenocysteine-containing protein SELENOI (SelI) in tumor cell lines. Dokl. Biochem. Biophys. 2017, 476, 320–322. [Google Scholar] [CrossRef] [PubMed]
  109. Lee, D.; Xu, I.M.-J.; Chiu, D.K.-C.; Leibold, J.; Tse, A.P.-W.; Bao, M.H.-R.; Yuen, V.W.-H.; Chan, C.Y.-K.; Lai, R.K.-H.; Chin, D.W.-C.; et al. Induction of oxidative stress through inhibition of thioredoxin reductase 1 is an effective therapeutic approach for hepatocellular carcinoma. Hepatology 2019, 69, 1768–1786. [Google Scholar] [CrossRef] [PubMed]
  110. Lencioni, R.; de Baere, T.; Soulen, M.C.; Rilling, W.S.; Geschwind, J.F.H. Lipiodol transarterial chemoembolization for hepatocellular carcinoma: A systematic review of efficacy and safety data. Hepatology 2016, 64, 106–116. [Google Scholar] [CrossRef]
  111. Liu, Q.; Qian, Y.; Li, P.; Zhang, S.; Liu, J.; Sun, X.; Fulham, M.; Feng, D.; Huang, G.; Lu, W.; et al. (131)I-Labeled Copper Sulfide-Loaded Microspheres to Treat Hepatic Tumors via Hepatic Artery Embolization. Theranostics 2018, 8, 785–799. [Google Scholar] [CrossRef]
  112. Lewis, A.L.; Gonzalez, M.V.; Lloyd, A.W.; Hall, B.; Tang, Y.; Willis, S.L.; Leppard, S.W.; Wolfenden, L.C.; Palmer, R.R.; Stratford, P.W. DC bead: In vitro characterization of a drug-delivery device for transarterial chemoembolization. J. Vasc. Interv. Radiol. JVIR 2006, 17 Pt 1, 335–342. [Google Scholar] [CrossRef]
  113. Kudo, M.; Finn, R.S.; Qin, S.; Han, K.H.; Ikeda, K.; Piscaglia, F.; Baron, A.; Park, J.W.; Han, G.; Jassem, J.; et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial. Lancet 2018, 391, 1163–1173. [Google Scholar] [CrossRef]
  114. Chung, Y.H.; Han, G.; Yoon, J.H.; Yang, J.; Wang, J.; Shao, G.L.; Kim, B.I.; Lee, T.Y.; Chao, Y. Interim analysis of START: Study in Asia of the combination of TACE (transcatheter arterial chemoembolization) with sorafenib in patients with hepatocellular carcinoma trial. Int. J. Cancer 2013, 132, 2448–2458. [Google Scholar] [CrossRef]
  115. Llovet, J.M.; Ricci, S.; Mazzaferro, V.; Hilgard, P.; Gane, E.; Blanc, J.F.; de Oliveira, A.C.; Santoro, A.; Raoul, J.L.; Forner, A.; et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 2008, 359, 378–390. [Google Scholar] [CrossRef]
  116. Liu, L.; Cao, Y.; Chen, C.; Zhang, X.; McNabola, A.; Wilkie, D.; Wilhelm, S.; Lynch, M.; Carter, C. Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 2006, 66, 11851–11858. [Google Scholar] [CrossRef]
  117. Meyer, T.; Fox, R.; Ma, Y.T.; Ross, P.J.; James, M.W.; Sturgess, R.; Stubbs, C.; Stocken, D.D.; Wall, L.; Watkinson, A.; et al. Sorafenib in combination with transarterial chemoembolisation in patients with unresectable hepatocellular carcinoma (TACE 2): A randomised placebo-controlled, double-blind, phase 3 trial. The lancet. Gastroenterol. Hepatol. 2017, 2, 565–575. [Google Scholar] [CrossRef]
  118. Kroschinsky, F.; Stolzel, F.; von Bonin, S.; Beutel, G.; Kochanek, M.; Kiehl, M.; Schellongowski, P. New drugs, new toxicities: Severe side effects of modern targeted and immunotherapy of cancer and their management. Crit. Care 2017, 21, 89. [Google Scholar] [CrossRef]
  119. Hutson, T.E.; Al-Shukri, S.; Stus, V.P.; Lipatov, O.N.; Shparyk, Y.; Bair, A.H.; Rosbrook, B.; Andrews, G.I.; Vogelzang, N.J. Axitinib versus sorafenib in first-line metastatic renal cell carcinoma: Overall survival from a randomized phase III trial. Clin. Genitourin. Cancer 2017, 15, 72–76. [Google Scholar] [CrossRef]
  120. Zheng, L.; Li, C.; Huang, X.; Lin, X.; Lin, W.; Yang, F.; Chen, T. Thermosensitive hydrogels for sustained-release of sorafenib and selenium nanoparticles for localized synergistic chemoradiotherapy. Biomaterials 2019, 216, 119220. [Google Scholar] [CrossRef] [PubMed]
  121. Al-Noshokaty, T.M.; Mesbah, N.M.; Abo-Elmatty, D.M.; Abulsoud, A.I.; Abdel-Hamed, A.R. Selenium nanoparticles overcomes sorafenib resistance in thioacetamide induced hepatocellular carcinoma in rats by modulation of mTOR, NF-κB pathways and LncRNA-AF085935/GPC3 axis. Life Sci. 2022, 303, 120675. [Google Scholar] [CrossRef]
  122. Wu, D.; Wang, H.; Hou, X.; Chen, H.; Ma, Y.; Hou, Y.; Hong, J. Effects of gold core size on regulating the performance of doxorubicin-conjugated gold nanoparticles. Nano Res. 2018, 11, 3396–3410. [Google Scholar] [CrossRef]
  123. Zhang, Z.; Wang, X.; Li, B.; Hou, Y.; Yang, J.; Yi, L. Development of a novel morphological paclitaxel-loaded PLGA microspheres for effective cancer therapy: In vitro and in vivo evaluations. Drug Deliv. 2018, 25, 166–177. [Google Scholar] [CrossRef]
  124. Acharya, S.; Nithyananthan, S.; Thirunavukkarasu, C. Selenium Nanoparticles Show Anticancer Activity Through Regulation of HIF-1α and HIF-2α Under Hypoxic Condition in Liver Cancer Cells. DNA Cell Biol. 2023, 42, 433–444. [Google Scholar] [CrossRef]
Figure 1. Activation of various signaling pathways leading to apoptosis of liver cancer cells after their treatment with SeNPs of various origins. Microsoft Paint was used to create the figure.
Figure 1. Activation of various signaling pathways leading to apoptosis of liver cancer cells after their treatment with SeNPs of various origins. Microsoft Paint was used to create the figure.
Cells 13 01102 g001
Figure 2. Activation of various signaling pathways leading to apoptosis and autophagy of liver cancer cells after their treatment with SeNPs of various origins. Microsoft Paint was used to create the figure.
Figure 2. Activation of various signaling pathways leading to apoptosis and autophagy of liver cancer cells after their treatment with SeNPs of various origins. Microsoft Paint was used to create the figure.
Cells 13 01102 g002
Table 1. Molecular mechanisms activated by various SeNPs in in vitro and in vivo HCC models.
Table 1. Molecular mechanisms activated by various SeNPs in in vitro and in vivo HCC models.
Nanoparticle CompositionObject of StudyMolecular Mechanisms Activated by NanoparticlesForm of Cell DeathRef.
SeNPs or SeSo
Selenium nanoparticles
or
sorafenib + selenium nanoparticles
HepG2 cellsPromote an increase in the expression of a number of pro-apoptotic genes, including GADD34, BAK, BAX, PUMA, CASP-3 and CASP-4; activate ER stress through the PERK signaling pathway; cause dose-dependent generation of various calcium signalsApoptosis[45]
Cur-SeNPs
Curcumin + selenium nanoparticles
HepG2 cells Activate the PI3K/Akt/mTOR pathwayApoptosis[27]
siRNA-PEI-SeNPs
Small interfering RNA + polyethylenimine + selenium nanoparticles
HepG2 cellsReduce the expression of HSP70; increase the activity of CASP-3 and the cleavage PARPApoptosis[48]
LP-SeNPs
Laminarin + selenium nanoparticles
HepG2 cellsIncrease BAX mRNA expression and CASP-9 cleavage; decrease BCL-2 levelsApoptosis[49]
HE-SeNPs
Hawthorn fruit extract + selenium nanoparticles
HepG2 cellsIncrease the level of CASP-9; decrease the level of BCL-2; induce intracellular oxidative stress and mitochondrial dysfunctionApoptosis[50]
BFP-SeNPs
Triple-helix β-glucan + selenium nanoparticles
HepG2 cellsInhibit cell proliferation through cell cycle arrest in the S phase; cause condensation of nuclear chromatin and severe nuclear shrinkage; increase ROS; decrease mitochondrial membrane potentialApoptosis[51]
siRNA-RGDfC-SeNPs
Small interfering RNA + peptide + selenium nanoparticles
HepG2 cellsKD of Oct4, which is accompanied by a decrease in the expression of Sox-2, Nanog, β-catenin and GSK-3β; activate Wnt/β-catenin signaling; reduce the expression of mTOR, AKT and PI3K; activate LC3-II and p62Autophagy[86]
QCT-SeNPs
Quercetin + selenium nanoparticles
Rat model of TAA-induced HCCIncrease oxidative stress; dysregulate the oncogenic p53/β-catenin/cyclin D signaling pathwayApoptosis or autophagy[39]
So + SeNPs
Sorafenib + selenium nanoparticles or sorafenib + selenium nanoparticles + radiation
HepG2 cells or mice with HCCReduce the expression of CD34 and Ki67; increase the growth of CASP-3. With radiation, hydrogel led to improved protein, se-rum creatinine, cholesterol and blood glucose levels.Apoptosis[120]
So + SeNPs
Sorafenib + selenium nanoparticles
Mouse model of TAA-induced HCCDecrease angiogenesis and metastasis, affecting the mTOR and NF-kB pathwaysApoptosis[121]
DOX + HA-SeNPs
Doxorubicin + hyaluronic acid + selenium nanoparticles
HepG2 cellsSuppress proliferation; promote the production of ROS; induce apoptosis through activation of CASP-3 pathwaysApoptosis[124]
Akt—protein kinase B; BAK—Bcl-2 homologous antagonist/killer; BAX—Bcl-2-like protein 4; Bcl-2—B-cell lymphoma 2; CASP—caspase; CD34—transmembrane phosphoglycoprotein encoded by the CD34 gene; GADD34—growth arrest and DNA damage gene; GSK-3β—glycogen synthase kinase-3 beta; KD—knockdown; Ki67—antigen Kiel 67; LC3-II—autophagosomal marker; mTOR—mammalian target of rapamycin; NF-kB—nuclear factor kappa light-chain enhancer of activated B cells; Oct4—octamer-binding transcription factor 4; p53—tumor protein P53; PEI—polyethylenimine; PERK—protein kinase RNA-like endoplasmic reticulum kinase; PI3K—phosphoinositide 3-kinase; PUMA—P53 upregulated modulator of apoptosis; ROS—reactive oxygen species; TAA—thioacetamide; Wnt—created from the names wingless and Int-1.
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

Varlamova, E.G. Molecular Mechanisms of the Therapeutic Effect of Selenium Nanoparticles in Hepatocellular Carcinoma. Cells 2024, 13, 1102. https://doi.org/10.3390/cells13131102

AMA Style

Varlamova EG. Molecular Mechanisms of the Therapeutic Effect of Selenium Nanoparticles in Hepatocellular Carcinoma. Cells. 2024; 13(13):1102. https://doi.org/10.3390/cells13131102

Chicago/Turabian Style

Varlamova, Elena G. 2024. "Molecular Mechanisms of the Therapeutic Effect of Selenium Nanoparticles in Hepatocellular Carcinoma" Cells 13, no. 13: 1102. https://doi.org/10.3390/cells13131102

APA Style

Varlamova, E. G. (2024). Molecular Mechanisms of the Therapeutic Effect of Selenium Nanoparticles in Hepatocellular Carcinoma. Cells, 13(13), 1102. https://doi.org/10.3390/cells13131102

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