Next Article in Journal
Review and Analysis of Heat Transfer in Spacer-Filled Channels of Membrane Distillation Systems
Next Article in Special Issue
Electric Fields at the Lipid Membrane Interface
Previous Article in Journal
Innovative Bioactive Nanofibrous Materials Combining Medicinal and Aromatic Plant Extracts and Electrospinning Method
Previous Article in Special Issue
The Membrane-Mediated Interaction of Liquid-Ordered Lipid Domains in the Presence of Amphipathic Peptides
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toxic Effects of Penetrating Cations

1
Department of Molecular Energetics of Microorganisms, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40 Leninskie Gory, 119991 Moscow, Russia
2
Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Premises 8, Bldg. 1, Village of Institute of Poliomyelitis, Settlement “Moskovskiy”, 108819 Moscow, Russia
3
Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 LeninskiyProspekt, 119071 Moscow, Russia
*
Author to whom correspondence should be addressed.
Membranes 2023, 13(10), 841; https://doi.org/10.3390/membranes13100841
Submission received: 21 August 2023 / Revised: 8 October 2023 / Accepted: 19 October 2023 / Published: 22 October 2023

Abstract

:
As mitochondria are negatively charged organelles, penetrating cations are used as parts of chimeric molecules to deliver specific compounds into mitochondria. In other words, they are used as electrophilic carriers for such chemical moieties as antioxidants, dyes, etc., to transfer them inside mitochondria. However, unmodified penetrating cations affect different aspects of cellular physiology as well. In this review, we have attempted to summarise the data about the side effects of commonly used natural (e.g., berberine) and artificial (e.g., tetraphenylphosphonium, rhodamine, methylene blue) penetrating cations on cellular physiology. For instance, it was shown that such types of molecules can (1) facilitate proton transport across membranes; (2) react with redox groups of the respiratory chain; (3) induce DNA damage; (4) interfere with pleiotropic drug resistance; (5) disturb membrane integrity; and (6) inhibit enzymes. Also, the products of the biodegradation of penetrating cations can be toxic. As penetrating cations accumulate in mitochondria, their toxicity is mostly due to mitochondrial damage. Mitochondria from certain types of cancer cells appear to be especially sensitive to penetrating cations. Here, we discuss the molecular mechanisms of the toxic effects and the anti-cancer activity of penetrating cations.

1. Introduction

There are two ways for lipophilic cations to cross a phospholipid bilayer. Single molecules of cations with small charged moieties (e.g., benzalkonium) cannot penetrate the bilayer because, in the aqueous phase, the moiety is attached to a number of water molecules via hydrogen bonds. Thus, such an aggregate cannot penetrate between the phospholipid molecules for sterical reasons. Additionally, the electrostatic Born energy of an ion in a dielectric medium (membrane) scales as the inverse ion radius [1]; for this reason, smaller ions have higher electrostatic energy inside the membrane as compared to larger ones, although for non-spherical molecules the membrane permeability strongly depends on the details of their molecular shape [2]. Still, such lipophilic molecules can cross the membrane via transient pores. If the effective cross-section of the polar parts of the ions is larger than the cross-section of their hydrophobic tail (that is usually the case), they locally induce a convex patch in the membrane monolayer, i.e., induce a positive spontaneous curvature [3,4]. Such curvature generally favours the formation of pores [5], and such cations can act as detergents. If the cation surface concentration increases, the curvature of the monolayer changes, leading to the formation of the pore [6]. Next, the molecules diffuse to the opposite monolayer, and the pore closes spontaneously. A similar effect was observed for the amphipathic peptide PGLa [7]. Upon incorporation into the outer monolayer of the membrane of giant unilamellar vesicles (GUV), this peptide makes the membrane locally convex. An increase in the peptide concentration leads to the formation of small transient pores, via which the peptide passes to the inner monolayer of the GUV membrane [7].
An alternative way is to cross without disrupting the integrity of the lipid bilayer. The lipocations, which lack the voluminous layer of hydrogen bond-attached water molecules, are capable of this. To avoid the formation of the halo of water molecules, the charge has to be either masked by hydrophobic groups, or delocalized, or both (e.g., triphenylphosphonium (TPP) and rhodamine, respectively, Figure 1). The latter group of lipocations is likely to be less harmful for the biological membranes than the former one. Indeed, such lipocations do not make pores in the membrane, which depolarise the cell plasma membrane and enable the release of the cytosolic constituents. The chemical formulas of the most well-known penetrating cations of this group are shown in Figure 1. These lipocations, or their derivatives (e.g., alkylated forms, dimers, etc.) are being widely used to deliver antioxidants and other types of chemical moieties to the mitochondria of human cells to improve their physiology. Nevertheless, even the molecules of this type of lipocations could be toxic for cells. In our review, we focus on the side effects of this type of electrophilic carrier.

1.1. Triphenyl Phosphonium

Tetraphenylphosphonium and the alkylated derivatives of triphenylphosphonium are probably the most well-studied artificial penetrating cations. Their toxic effects were studied in a variety of experimental systems. When alkylated derivatives of TPP are added to mitochondria, they stimulate respiration. This effect is due to the temporary depolarization of mitochondria due to the passage of a charged molecule through the phospholipid bilayer. Moreover, alkyl-TPP can stimulate the protonophore activity of free fatty acids [8]. At the same time, high concentrations inhibit respiration. That could be due to the direct inhibition of respiratory enzymes or mediation by their detergent action on the mitochondrial inner membrane (see review [9]).At the level of whole cells, we have shown that one of the TPP derivatives, C12TPP, simultaneously induces the expression of multidrug resistance pump (MDR) proteins and also inhibits their activity. The inhibition is not toxic for yeast cells, but the inhibition might be used to enhance the effect of antimycotic drugs which are MDR substrates [10,11]. TPP has been shown to cause hepatotoxicity, but the cellular mechanisms are still not clear. Probably, the liver damage is due to apoptotic cell death because it causes apoptosis in the cultured hepatocyte AML12 cell line [12]. Additionally, one can hypothesise that the apoptosis of liver cells induced by TPP and similar large-headed lipocations is due to the disturbance of cell signalling. It has been shown that several gangliosides with large polar heads and regular lipid tails can induce apoptosis in T-lymphocytes [13,14]. Gangliosides are also known for their ability to alter the ensemble properties of small, ordered lipid–protein domains of plasma membranes [15]; such domains are also called rafts [16]. Similar effects have recently been observed for lysolipids, also with relatively large polar heads and small lipid tails [17]. This result points out that the modification of the raft ensemble properties is not specific to the chemical structure of the particular lipid, e.g., gangliosides, lysolipids. Therefore, TPP might act in a similar manner. Moreover, various cell receptors, and especially death receptors, are raft-associated proteins [18]. Thus, by modifying raft properties, large-headed lipids are able to induce apoptosis even in the absence of specific ligands of death receptors. Thus, the induction of apoptosis and, more generally, the influence on the raft ensemble leading to disturbance of normal cell signalling [19] are, possibly, two more side-effects of penetrating cations. Not surprisingly, when the excess of C12TPP is administered to mice, the reason for its toxic effect is also due to liver damage. It has been shown that it triggers massive cell death in liver zone 3. In fact, this is also not surprising, because such damage is typically induced by phospho-organic compounds [20]. The latter points out that, most likely, the toxicity is induced by the degradation products of C12TPP. It has also been shown that decyl-triphenylphosphonium can be used to selectively kill multiple myeloma cancer cells. Treatment with C10TPP increased intracellular steady-state pro-oxidant levels in stem-like and mature multiple myeloma cells. Ectopic expression of manganese superoxide dismutase is able to suppress the increases in oxidant production induced by C10TPP treatment [21], which also points at the prooxidant activity of C10TPP.Possibly, the anticancer effect of the TPP moiety contributes to the anticancer effect of TPP-linked compounds. Indeed, it is well known that TPP is used to deliver a number of drugs to the mitochondria of tumour cells in order to kill them or inhibit their proliferation. Interestingly, TPP-based mitochondria-targeted anticancer drugs mainly target tumour cells with high membrane potential. Possibly, the latter is due to an increased accumulation of the positively charged compounds in hyper-polarized mitochondria [22].

1.2. Dequalinium Chloride (DQ)

Dequalinium is a quaternary ammonium compound with well-documented antibacterial activity. It is being used to treat several types of infections. While being a penetrating cation, it is also capable of lysing bacterial outer membranes [23]. Its targets within the bacterial cells include inhibition of glycolysis, inhibition of F1-ATPase, and protein biosynthesis via interfering with ribosomal activity [23,24]. Similar to the alkylated forms of triphenylphosphonium, DQ does display selective toxicity for cancer cells. The anticancer activity of DQ is due to its ability to target mitochondria, inhibit certain kinases, and affect Ca2+-activated K+ channels. It specifically targets the mitochondrial membrane of epithelial carcinoma cells, leading to the disruption of cellular energy production. This disruption leads to oxidative stress by reducing glutathione levels and increasing reactive oxygen species levels in a cell-specific manner [25]. DQ also interferes with signalling pathways, such as by downregulating the Raf/MEK/ERK1/2 and PI3K/Akt pathways in NB4 leukaemia cells [26,27,28]. The mitochondrial damage induced by DQ is an early event in its mechanism of action [26]. This damage includes the depletion of mitochondrial DNA [29]. DQ also causes a decrease in mitochondrial membrane potential; it also triggers mitochondrial ROS generation and the depletion of ATP [30].

1.3. Pyrvinium and Cyanine

Pyrvinium, a penetrating cation related to cyanine dye, has been used for many decades as an anthelmintic. Similar to other clinically used penetrating cations, its mitochondrial localization and targeting are well documented [31]. In particular, it has been shown that pyrvinium inhibits mitochondrial respiration in parasitic worms [32]. Its mitochondrial accumulation is also responsible for its activity against pathogenic fungi such as Candida auris [33] and Aspergillus fumigatus [34].
Over the past two decades, increasing evidence has emerged showing pyrvinium to be a strong selective anti-cancer molecule in various human cancers in vitro and in vivo [31]. Similar to its anti-parasitic activity, the anti-cancer action of pyrvinium relies on the inhibition of mitochondrial function. There is strong evidence that the inhibition of respiratory complex I is largely responsible for the anticancer activity. Indeed, it was reported that the inhibitory concentrations are in the sub-micromolar range [35,36]. At the same time, it has been noticed that cancer cell lines with depleted mitochondrial DNA are resistant to pyrvinium [35,37]. Complex I is not the only mitochondrial target of pyrvinium. Fumarate reductase [38] and mitochondrial DNA [39] were reported as others. The latter is due to the binding of pyrvinium to G-quadruplex structures, which are more frequent in the mitochondrial DNA than in the nuclear one [40,41]. Mitochondria are not the only targets of pyrvinium. In the cytosol, it binds and activates Casein kinase 1α, leading to the degradation of beta-catenin, the key effector of the WNT signalling pathway. This also contributes to the anticancer activity of pyrvinium [42,43,44]. It also binds and inhibits a number of other non-mitochondrial proteins, including ELAVL1/HuR, a post-transcriptional regulator of a set of genes driving the survival of cancer cells [45,46,47] and the androgen receptor. The latter property has been explored to treat prostate cancer [48,49,50].
Cyanine fluorophores have a high extinction coefficient and relatively long absorption and emission wavelengths. Combined with their low toxicity in biological samples, cyanine fluorophores are promising candidates for biomedical applications [51]. Cyanine fluorophores are used for photodynamic therapy and photothermal therapy, which, due to the high precision of localization of the effect, are very promising methods for treating cancer [52].

1.4. Rhodamine

Rhodamine is a fluorescent dye commonly used in laboratory practice to visualise mitochondria. Despite its affinity for mitochondria, its toxicity is attributed to the inhibition of cytosolic antioxidant enzymes, superoxide dismutase (SOD), catalase (CAT), and guaiacol peroxidase (GPOD). It is believed that the inhibition is caused by the degradation products of rhodamine, with degradation being caused by cytochrome P450 [53,54]. Similar to the aforementioned penetrating lipocations, Rhodamine is specifically toxic for carcinoma cancer cells [55]. It has been suggested that the reason for the selective toxicity is due to the fact that these cancer cells have a higher electric potential at their plasma membranes than the normal ones, leading to an increased accumulation of the compound within the cells [56]. However, it should be mentioned that not all cancer types have an increase in plasma membrane (PM) transmembrane potential. Nevertheless, rhodamine was accumulating inside the mitochondria of the carcinoma cells, and its re-localisation to the cytosol was observed shortly before the cell death [55]. This observation suggests that mitochondrial damage is the primary reason for the toxicity of rhodamine.
In addition, rhodamine-based fluorescent labels were shown to induce lipid oxidation that leads, in particular, to an alteration of the dynamics of membrane rafts [57], potentially yielding perturbation of cell signalling, although the mechanical properties of membranes remained almost unaltered [58,59].
Interestingly, it has been reported that certain rhodamine conjugates do possess surfactant activity [60]. Thismeans that, possibly, the penetration of such large molecules through the membrane is accompanied by the disturbance of the integrity of the phospholipid bilayer. Thus, according to our definition of the penetrating cation (see above), it is likely that these conjugates are not true penetrating cations. The abovemay also explain the mitochondrial damage induced by various rhodamines.

1.5. Berberine, Palmatine and Sanguinarine

Berberine, palmatine and sanguinarine are chemically related compounds produced by plants as a defence against various parasites. Not surprisingly, they have several intracellular targets and affect cellular physiology by a number of independent means. First, berberine and sanguinarine bind DNA and inhibit DNA synthesis [61]. Palmatine was also shown to specifically induce DNA strand breakage in cultured cancer cells [62]. Isoquinoline alkaloids can interact with DNA in a variety of ways: by binding to phosphate groups, or to a small groove, or as intercalators [63]. Possibly, that explains why berberine, palmatine and sanguinarine target DNA. Berberine, by binding to histone-DNA complexes, can interfere with vital cellular processes such as cell division and activate apoptosis in living cells, leading to the selective death of cancer cells [64,65].
Apart from binding DNA, these compounds interact with and inhibit several extra- and intracellular proteins. Acetylcholine esterase, butyrylcholinesterase, and choline acetyltransferase are inhibited by them, as are the receptors alpha 1- and alpha 2-adrenergic, nicotinergic, muscarinergic, and serotonin2 receptors [61].
Sanguinarine [13-methyl(1,3)benzodioxolo(5,6-c)-1,3-dioxolo(4,5)phenanthridinium] is more toxic than berberine or palmatine. The additional toxicity of sanguinarine is mostly due to its inhibition of the Na+-K+-ATPase transmembrane protein [63]. Sanguinarine has also been shown to cause cell membrane damage by lipid peroxidation by free radicals, including ROS and reactive nitrogen species (RNS). It also possesses DNA polymerase inhibition activity and causes the accumulation of pyruvate due to increased glycogenolysis [66].

1.6. E-4-(1H-indol-3-ylvinyl)-N-methylpyridinium Iodide (F16)

A novel penetrating cation, F16 has been recently shown to possess uncoupling activity. It was also reported that it specifically accumulates in the mitochondria of carcinoma cells and inhibits their growth. It has been concluded that mitochondrial toxicity by the means of uncoupling mediates the anticancer activity of F16 [67]. F16 conjugates with ursolic, oleanolic, maslinic, and corosolic acids, which show increased toxicity to cancer cells. This toxicity is also due to an increase in ROS that occurs during the action of the conjugates on mitochondria [68]. Work is underway to study the anticancer toxicity of F16 derivatives; for example, it has been shown that 5-Br-7MeF16 (5BMF) has a low IC50 of ∼50 nM (to H2228 cells) and a high cancer to normal cell selectivity index of 225 [69].

1.7. Methylene Blue

Methylene blue (MB) induces non-toxic hydrogen peroxide production and, in this way, protects mitochondria, in particular, from rotenone toxicity. For this reason, methylene blue displays a broad range of neuroprotective activity. Its therapeutic effect is also due to the activation of mitochondrial biogenesis, the latter being mediated by the Nrf2/ARE signalling cascade. It is believed that the cascade is triggered by H2O2 generation induced by methylene blue [70,71]. Interestingly, similar to the other penetrating cations, methylene blue is selectively toxic to cancer cells. It was shown that lung cancer cells treated with methylene blue show reduced activity of Hsp70, the protein that is essential for their survival [72]. It is not clear whether this is a direct inhibition or a consequence of mitochondrial damage or H2O2 production. MB is able to disrupt NO production by inhibiting guanylyl cyclase and endothelial NO synthase [73]. MB can accumulate in the mitochondria and lysosomes of HaCaT cells and demonstrate phototoxicity [74].

1.8. Sepantronium Bromide (Ym155)

In preclinical studies, the imidazolium-based compound Ym155 displayed selective suppression of the growth of many types of cancer cell lines. It was discovered as a result of screening for inhibitors of survivin expression. However, its biomolecular target remains unknown [75]. The reason for that has started to become clear only recently. It has been shown that, being a penetrating cation, it accumulates in mitochondria and binds to mitochondrial DNA, causing energy depletion in the cells [76]. Ym155 generates ROS in mitochondria, causing DNA damage and inhibiting survivin. This mediates the cytotoxic effects of Ym155 on triple-negative breast cancer TNBC cells through both apoptotic and necrotic pathways [77].

1.8.1. Metformin and Doxorubicin

The compounds metformin and doxycycline differ in their way of penetration into the cells from other chemicals discussed in the review. Nevertheless, their therapeutic effects are similar to those of the other penetrating cations.

1.8.2. Metformin

Metformin, a derivative of guanidine, has been used for decades in the treatment of type 2 diabetes. Metformin has a pKa of 12.4 and thus, under physiological conditions, it exists in a protonated form. It has been reported that metformin enters cells via organic cation transporters (Octs) [78] and then targets mitochondria. For this reason, mitochondria play a key role in both pharmacological and toxic actions of metformin [79]. A number of studies have shown that metformin selectively inhibits the growth of several types of cancer cells [80]. It has been suggested that the primary reason for the anticancer activity of metformin is inhibition of mitochondrial energy production [80]. The latter is consistent with the observation that metformin inhibits complex I in the mitochondrial respiratory chain [79]. Metformin has also been shown to induce ROS formation, thus stimulating redox signalling mechanisms [80]. It has been suggested that this is due to complex I inhibition [81]. Formation of ROS may be a negative side-effect of this cation if it occurs in normal cells; it was shown that agents that depolarize the mitochondrial membrane and prevent ROS formation in cells under oxidative stress-associated conditions exhibit neuroprotective activity [82,83]. There is also evidence that metformin directly activates AMP-dependent protein kinase (AMPK), the key sensor of cellular energy status, via the lysosomal AXIN-dependent pathway [84], leading to the suppression of cell growth. It has also been reported that metformin inhibits PI3K/AKT signalling, the pathway which is essential for cell growth and proliferation [80]. Interestingly, it has been shown that metformin induces apoptosis in ovarian cancer cells by changing the balance of Bax and Bcl-2 proteins in an AMPK-independent manner [85]. Another target of metformin is DNA methylation. It has been reported that the addition of metformin leads to a decrease in S-adenosylmethionine levels in C. elegans, which forces the H3K4 methyltransferase/demethylase complex to downregulate the targets [86]. To summarise, while mitochondria are the primary target of metformin, the compound also affects a set of non-mitochondrial targets within a cell.

1.8.3. Doxorubicin (Dox)

Doxorubicin (Dox) is an antibiotic, which is produced by Streptomyces peucetius bacterium. pKa of Dox equals 8.01, so most Dox molecules are charged at physiological pH [87]. To our knowledge, it has not been shown directly that Dox in protonated form is capable of crossing the lipid bilayers. Still, it has been shown that, when added to MDA-MB-435 cancer cells, it co-localizes with mitochondria [88]. Its ability to selectively damage mitochondria (see below) also points out that protonated Dox is a penetrating cation.
Dox is commonly used to treat different cancers. It has been reported that Dox-induced killing of cancer cells is mediated by intercalation within DNA base pairs, preventing the synthesis of both DNA and RNA and inhibiting topoisomerase II. Dox also intercalates mitochondrial DNA [89]. This can cause breaks in the mitochondrial DNA, either in one strand or both. Moreover, doxorubicin combined with iron also causes damage to DNA through the production of harmful ROS [90,91,92].
There was an attempt to increase the mitochondrial accumulation of Dox. For this purpose, the molecule of Dox was linked to TPP. The attempt was obviously successful. It was demonstrated that, unlike Dox, chimeric Dox-TPP accumulated not only in the mitochondria of cancerous MDA-MB-435 cells, but also in mitochondria of MDA-MB-435 wild-type cells, the ones with lower mitochondrial membrane potential [88]. Later, it was shown that Dox-TPP-containing nanoparticles of various types have a great potential to selectively eliminate cancer cells by targeting their mitochondria [93,94,95].
Treatment with Dox can increase the risk of death for cancer patients because of its cardiotoxicity. Cardiotoxicity can lead to the development of cardiomyopathy and, ultimately, congestive heart failure. Again, one of the mechanisms responsible for Dox-induced cardiotoxicity is impaired mitochondrial function and ROS generation in heart cells. It has been demonstrated that mitochondria are one of the major sites for Dox-induced oxidative stress and cellular damage [96].
Dox-induced ROS generation activates four proteolytic systems: (1) the ubiquitin proteasome system (UPS), (2) calpain, (3) caspase-3, and (4) autophagy. (1) Polyubiquitinated damaged proteins are degraded by UPS. It has been shown that treatment with Dox can increase this protein breakdown in cardiac muscle, which can ultimately lead to damage in the heart [97]. (2) Altered levels of Ca2+ are also one of the mechanisms involved in Dox-induced cardiotoxicity. Dox treatment causes calcium overload, which increases the activity of a protein calpain. This protein cleaves cytoplasmic and nuclear substrates, leading to apoptosis [98]. (3) Caspases are enzymes that play a role in programmed cell death. Studies have shown that treatment with Dox can activate the apoptosis regulators, resulting in the activation of caspase-3 in cardiac muscle, leading to cell death [99]. Consistent with this, inhibiting caspase-3 activity can help to reduce the cardiotoxiceffects of Dox [100]. (4) The level of autophagy is low in the heart muscle but, under stress, autophagy is increased as a response [101]. Dox treatment increases the Beclin-1/Bcl-2 ratio, which is responsible for activation of both autophagy and apoptosis [89].

2. Discussion

Table 1 presents the summary of known effects of the penetrating cations. The table shows that their only common features are the disruption of phospholipid bilayers and the selective toxicity towards cancer cells.
The feature not shown in the table is that all of the compounds inflict damage on mitochondria. Although it is apparent that small lipophilic cations are mitochondrial toxins, the exact basis for their toxicity is difficult to pinpoint. It has been noticed that the relatively hydrophobic compounds (Rh123) affect electron transport and ATP synthesis, while the relatively hydrophilic ones perturb the matrix proteins and also inflict mito-DNA damage ([102,103,104,105,106] and Table 1). One can speculate that the membranophilic nature of the cations will also affect the phospholipids of the mitochondrial inner membrane. It is well known that typical lipids of biological membranes have conical (small polar head and large hydrophobic tails) or cylindrical (approximately equal cross-sections of polar head and hydrophobic tails) molecular shapes. At the same time, they have negative or zero spontaneous curvature [107,108]. It has been reported that such molecular shapes prevent the spontaneous formation of pores in the planar membranes [5,109]. On the contrary, membranes containing molecules having an inverse conical shape (large polar head, small hydrophobic tail) are prone to forming pores [110]. The polar groups of lipophilic cations shown in Figure 1 are likely to have an affinity for the polar heads of the lipid bilayer. Indeed, hydrophobic parts of the cations tend to avoid contact with water and accumulate inside the hydrophobic membrane core with their aromatic groups being mostly distributed to the polar/hydrophobic interface of a lipid monolayer. Each of the cations shown in Figure 1 has relatively small hydrophobic parts and large polar or/and aromatic parts. Upon incorporation into the membrane, they, obviously, disturb the ordering of the phospholipid molecules, and this disturbance might cause an increase in the permeability of various small molecules through the membrane either via packing defects or by pore formation. In fact, non-penetrating cations, such as benzalkonium, disturb the lipid packing in a cholesterol-sensitive manner [111] and inflict the same type of damage on the lipid bilayers as the penetrating cations. This property makes them powerful antibacterial agents. Indeed, while the outer membrane of eukaryotic cells is protected by a high content of sterols, bacterial membranes typically lack sterols and thus are vulnerable to the disruption of ordering of the phospholipid molecules. For example, imidazolium-based ionic liquids can be used as antibacterial agents. Our preliminary data indicate that they do not penetrate through the bilayer, and thus their antibacterial effect is mostly due to their membrane-disrupting action.
Another common feature of the discussed penetrating cations is their anticancer activity, especially against the carcinoma cells. This was noticed a couple of decades ago [25], and further studies confirmed this observation (see above). The reason for that might be due to the fact that typically, for energy supply, cancer cells rely on glycolysis (Warburg effect). This type of metabolism is typically accompanied by the inhibition of mitochondrial respiration. As a certain level of mitochondrial membrane potential is required for cell survival, such cells use mitochondrial ATP synthase in a reverse fashion for the potential generation. As this type of the potential generation is much less powerful than respiration, even a relatively minor increase in the proton conductivity of the mitochondrial inner membrane might be lethal for the cell. Such an increase might be induced by the accumulation of the lipophilic cations in the mitochondrial inner membrane. Indeed, yeast cells that lack functional mitochondrial DNA and rely solely on glycolysis are much more sensitive to conventional protonophores than those with intact mito-DNA [112]. The same is true for cancer cells: at least certain cancer cell lines are more sensitive to uncouplers than normal cells [113]. As all of the penetrating cations in one way or another decrease the membrane potential, this may explain their anti-cancer activity.
An alternative explanation of the anti-cancer activity of the penetrating cations is that many types of cancer cells display an elevated level of the membrane potential. Mitochondria of such cells possess fully functional respiratory chains and, at the same time, non-functional ATP synthase. The inhibition is due to the expression of a special inhibitory protein, IF1. Apparently, the increased charge of mitochondria presumes an increased level of penetrating cation accumulation. As some of them inhibit specific mitochondrial enzymes or bind mitochondrial DNA, such accumulation might render these types of cancer cells more sensitive to the cations than the normal ones.
Another possible reason for the sensitivity of cancer cells to the penetrating cations is that many types of cancer cells show elevated activity of multidrug resistance pumps (MDRs). The reason is that these pumps extrude various chemicals used for chemotherapy. We have recently shown that a set of penetrating cations, alkylrhodamines, are inhibitors of MDRs. In the case of yeast, they display strong synergy with antimycotic drugs [114]. Possibly, in the case of cancer cells the penetrating cations act in the same way.

3. Conclusions

To summarise, the analysis of the literature points out that the only common features of all the aforementioned penetrating cations are their anticancer activity and damage to mitochondria. While each of the cations damages mitochondria in its own way, they all have something in common in this respect, namely the disruption of the ordering of the phospholipid molecules of the inner mitochondrial membrane. This, most likely, contributes to their selective toxicity toward cancer cells. Indeed, it is well known that the depletion of energy (ATP) in cancer cells is a commonly used strategy to treat tumours (reviewed in [115,116]). While the reason for the increased sensitivity of the cancer cells to penetrating cations is not exactly clear, the very fact, in our opinion, is beyond doubt.

Author Contributions

A.Z. prepared the illustration. S.S., A.Z., S.A., D.K. and F.S. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Russian Science Foundation grant N 22-24-00533.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Born, M. Volumen Und Hydratationswärme Der Ionen. Z. Phys. 1920, 1, 45–48. [Google Scholar] [CrossRef]
  2. Ebert, A.; Hannesschlaeger, C.; Goss, K.-U.; Pohl, P. Passive Permeability of Planar Lipid Bilayers to Organic Anions. Biophys. J. 2018, 115, 1931–1941. [Google Scholar] [CrossRef] [PubMed]
  3. Fuller, N.; Rand, R.P. The Influence of Lysolipids on the Spontaneous Curvature and Bending Elasticity of Phospholipid Membranes. Biophys. J. 2001, 81, 243–254. [Google Scholar] [CrossRef]
  4. Dymond, M.K. Lipid Monolayer Spontaneous Curvatures: A Collection of Published Values. Chem. Phys. Lipids 2021, 239, 105117. [Google Scholar] [CrossRef]
  5. Tazawa, K.; Yamazaki, M. Effect of Monolayer Spontaneous Curvature on Constant Tension-Induced Pore Formation in Lipid Bilayers. J. Chem. Phys. 2023, 158, 081101. [Google Scholar] [CrossRef]
  6. Huang, H.W. Molecular Mechanism of Antimicrobial Peptides: The Origin of Cooperativity. Biochim. Biophys. Acta 2006, 1758, 1292–1302. [Google Scholar] [CrossRef]
  7. Parvez, F.; Alam, J.M.; Dohra, H.; Yamazaki, M. Elementary Processes of Antimicrobial Peptide PGLa-Induced Pore Formation in Lipid Bilayers. Biochim. Biophys. Acta Biomembr. 2018, 1860, 2262–2271. [Google Scholar] [CrossRef]
  8. Severin, F.F.; Severina, I.I.; Antonenko, Y.N.; Rokitskaya, T.I.; Cherepanov, D.A.; Mokhova, E.N.; Vyssokikh, M.Y.; Pustovidko, A.V.; Markova, O.V.; Yaguzhinsky, L.S.; et al. Penetrating Cation/fatty Acid Anion Pair as a Mitochondria-Targeted Protonophore. Proc. Natl. Acad. Sci. USA 2010, 107, 663–668. [Google Scholar] [CrossRef] [PubMed]
  9. Zielonka, J.; Joseph, J.; Sikora, A.; Hardy, M.; Ouari, O.; Vasquez-Vivar, J.; Cheng, G.; Lopez, M.; Kalyanaraman, B. Mitochondria-Targeted Triphenylphosphonium-Based Compounds: Syntheses, Mechanisms of Action, and Therapeutic and Diagnostic Applications. Chem. Rev. 2017, 117, 10043–10120. [Google Scholar] [CrossRef] [PubMed]
  10. Knorre, D.A.; Markova, O.V.; Smirnova, E.A.; Karavaeva, I.E.; Sokolov, S.S.; Severin, F.F. Dodecyltriphenylphosphonium Inhibits Multiple Drug Resistance in the Yeast Saccharomyces Cerevisiae. Biochem. Biophys. Res. Commun. 2014, 450, 1481–1484. [Google Scholar] [CrossRef]
  11. Galkina, K.V.; Besedina, E.G.; Zinovkin, R.A.; Severin, F.F.; Knorre, D.A. Penetrating Cations Induce Pleiotropic Drug Resistance in Yeast. Sci. Rep. 2018, 8, 8131. [Google Scholar] [CrossRef] [PubMed]
  12. Li, M.; Liu, G.; Yuan, L.-X.; Yang, J.; Liu, J.; Li, Z.; Yang, C.; Wang, J. Triphenyl Phosphate (TPP) Promotes Hepatocyte Toxicity via Induction of Endoplasmic Reticulum Stress and Inhibition of Autophagy Flux. Sci. Total Environ. 2022, 840, 156461. [Google Scholar] [CrossRef] [PubMed]
  13. Molotkovskaya, I.M.; Kholodenko, R.V.; Zelenova, N.A.; Sapozhnikov, A.M.; Mikhalev, I.I.; Molotkovsky, J.G. Gangliosides Induce Cell Apoptosis in the Cytotoxic Line CTLL-2, but Not in the Promyelocyte Leukemia Cell Line HL-60. Membr. Cell Biol. 2000, 13, 811–822. [Google Scholar]
  14. Doronin, I.I.; Vishnyakova, P.A.; Kholodenko, I.V.; Ponomarev, E.D.; Ryazantsev, D.Y.; Molotkovskaya, I.M.; Kholodenko, R.V. Ganglioside GD2 in Reception and Transduction of Cell Death Signal in Tumor Cells. BMC Cancer 2014, 14, 295. [Google Scholar] [CrossRef]
  15. Galimzyanov, T.R.; Lyushnyak, A.S.; Aleksandrova, V.V.; Shilova, L.A.; Mikhalyov, I.I.; Molotkovskaya, I.M.; Akimov, S.A.; Batishchev, O.V. Line Activity of Ganglioside GM1 Regulates the Raft Size Distribution in a Cholesterol-Dependent Manner. Langmuir 2017, 33, 3517–3524. [Google Scholar] [CrossRef]
  16. Pike, L.J. Rafts Defined: A Report on the Keystone Symposium on Lipid Rafts and Cell Function. J. Lipid Res. 2006, 47, 1597–1598. [Google Scholar] [CrossRef] [PubMed]
  17. Krasnobaev, V.D.; Galimzyanov, T.R.; Akimov, S.A.; Batishchev, O.V. Lysolipids Regulate Raft Size Distribution. Front. Mol. Biosci. 2022, 9, 1021321. [Google Scholar] [CrossRef]
  18. Simons, K.; Toomre, D. Lipid Rafts and Signal Transduction. Nat. Rev. Mol. Cell Biol. 2000, 1, 31–39. [Google Scholar] [CrossRef]
  19. Li, B.; Qin, Y.; Yu, X.; Xu, X.; Yu, W. Lipid Raft Involvement in Signal Transduction in Cancer Cell Survival, Cell Death and Metastasis. Cell Prolif. 2022, 55, e13167. [Google Scholar] [CrossRef]
  20. Манских, В.Н. Патoмoрфoлoгия Лабoратoрнoй Мыши Рукoвoдствo в Трех Тoмах; ВАКО: Moscow, Russia, 2018. [Google Scholar]
  21. Schibler, J.; Tomanek-Chalkley, A.M.; Reedy, J.L.; Zhan, F.; Spitz, D.R.; Schultz, M.K.; Goel, A. Mitochondrial-Targeted Decyl-Triphenylphosphonium Enhances 2-Deoxy-D-Glucose Mediated Oxidative Stress and Clonogenic Killing of Multiple Myeloma Cells. PLoS ONE 2016, 11, e0167323. [Google Scholar] [CrossRef]
  22. Cheng, X.; Feng, D.; Lv, J.; Cui, X.; Wang, Y.; Wang, Q.; Zhang, L. Application Prospects of Triphenylphosphine-Based Mitochondria-Targeted Cancer Therapy. Cancers 2023, 15, 666. [Google Scholar] [CrossRef] [PubMed]
  23. Bailly, C. Medicinal Applications and Molecular Targets of Dequalinium Chloride. Biochem. Pharmacol. 2021, 186, 114467. [Google Scholar] [CrossRef]
  24. Mendling, W.; Weissenbacher, E.R.; Gerber, S.; Prasauskas, V.; Grob, P. Use of Locally Delivered Dequalinium Chloride in the Treatment of Vaginal Infections: A Review. Arch. Gynecol. Obstet. 2016, 293, 469–484. [Google Scholar] [CrossRef]
  25. Modica-Napolitano, J.S.; Aprille, J.R. Delocalized Lipophilic Cations Selectively Target the Mitochondria of Carcinoma Cells. Adv. Drug Deliv. Rev. 2001, 49, 63–70. [Google Scholar] [CrossRef]
  26. Sancho, P.; Galeano, E.; Estañ, M.C.; Gañán-Gómez, I.; Boyano-Adánez, M.D.C.; García-Pérez, A.I. Raf/MEK/ERK Signaling Inhibition Enhances the Ability of Dequalinium to Induce Apoptosis in the Human Leukemic Cell Line K562. Exp. Biol. Med. 2012, 237, 933–942. [Google Scholar] [CrossRef] [PubMed]
  27. García-Pérez, A.I.; Galeano, E.; Nieto, E.; Estañ, M.C.; Sancho, P. Dequalinium Induces Cytotoxicity in Human Leukemia NB4 Cells by Downregulation of Raf/MEK/ERK and PI3K/Akt Signaling Pathways and Potentiation of Specific Inhibitors of These Pathways. Leuk. Res. 2014, 38, 795–803. [Google Scholar] [CrossRef] [PubMed]
  28. Gañán-Gómez, I.; Estañ-Omaña, M.C.; Sancho, P.; Aller, P.; Boyano-Adánez, M.C. Mechanisms of Resistance to Apoptosis in the Human Acute Promyelocytic Leukemia Cell Line NB4. Ann. Hematol. 2015, 94, 379–392. [Google Scholar] [CrossRef]
  29. Schneider Berlin, K.R.; Ammini, C.V.; Rowe, T.C. Dequalinium Induces a Selective Depletion of Mitochondrial DNA from HeLa Human Cervical Carcinoma Cells. Exp. Cell Res. 1998, 245, 137–145. [Google Scholar] [CrossRef]
  30. Chan, C.F.; Lin-Shiau, S.Y. Suramin Prevents Cerebellar Granule Cell-Death Induced by Dequalinium. Neurochem. Int. 2001, 38, 135–143. [Google Scholar] [CrossRef]
  31. Schultz, C.W.; Nevler, A. Pyrvinium Pamoate: Past, Present, and Future as an Anti-Cancer Drug. Biomedicines 2022, 10, 3249. [Google Scholar] [CrossRef]
  32. Talaam, K.K.; Inaoka, D.K.; Hatta, T.; Tsubokawa, D.; Tsuji, N.; Wada, M.; Saimoto, H.; Kita, K.; Hamano, S. Mitochondria as a Potential Target for the Development of Prophylactic and Therapeutic Drugs against Schistosoma Mansoni Infection. Antimicrob. Agents Chemother. 2021, 65, e0041821. [Google Scholar] [CrossRef] [PubMed]
  33. Simm, C.; Weerasinghe, H.; Thomas, D.R.; Harrison, P.F.; Newton, H.J.; Beilharz, T.H.; Traven, A. Disruption of Iron Homeostasis and Mitochondrial Metabolism Are Promising Targets to Inhibit Candida Auris. Microbiol. Spectr. 2022, 10, e0010022. [Google Scholar] [CrossRef]
  34. Sun, Y.; Gao, L.; Zhang, Y.; Yang, J.; Zeng, T. Synergistic Effect of Pyrvinium Pamoate and Azoles Against Aspergillus Fumigatus in Vitro and in Vivo. Front. Microbiol. 2020, 11, 579362. [Google Scholar] [CrossRef] [PubMed]
  35. Harada, Y.; Ishii, I.; Hatake, K.; Kasahara, T. Pyrvinium Pamoate Inhibits Proliferation of Myeloma/erythroleukemia Cells by Suppressing Mitochondrial Respiratory Complex I and STAT3. Cancer Lett. 2012, 319, 83–88. [Google Scholar] [CrossRef]
  36. Xiao, M.; Zhang, L.; Zhou, Y.; Rajoria, P.; Wang, C. Pyrvinium Selectively Induces Apoptosis of Lymphoma Cells through Impairing Mitochondrial Functions and JAK2/STAT5. Biochem. Biophys. Res. Commun. 2016, 469, 716–722. [Google Scholar] [CrossRef]
  37. Xiang, W.; Cheong, J.K.; Ang, S.H.; Teo, B.; Xu, P.; Asari, K.; Sun, W.T.; Than, H.; Bunte, R.M.; Virshup, D.M.; et al. Pyrvinium Selectively Targets Blast Phase-Chronic Myeloid Leukemia through Inhibition of Mitochondrial Respiration. Oncotarget 2015, 6, 33769–33780. [Google Scholar] [CrossRef]
  38. Tomitsuka, E.; Kita, K.; Esumi, H. An Anticancer Agent, Pyrvinium Pamoate Inhibits the NADH-Fumarate Reductase System—A Unique Mitochondrial Energy Metabolism in Tumour Microenvironments. J. Biochem. 2012, 152, 171–183. [Google Scholar] [CrossRef] [PubMed]
  39. Schultz, C.W.; McCarthy, G.A.; Nerwal, T.; Nevler, A.; DuHadaway, J.B.; McCoy, M.D.; Jiang, W.; Brown, S.Z.; Goetz, A.; Jain, A.; et al. The FDA-Approved Anthelmintic Pyrvinium Pamoate Inhibits Pancreatic Cancer Cells in Nutrient-Depleted Conditions by Targeting the Mitochondria. Mol. Cancer Ther. 2021, 20, 2166–2176. [Google Scholar] [CrossRef]
  40. Falabella, M.; Fernandez, R.J.; Johnson, F.B.; Kaufman, B.A. Potential Roles for G-Quadruplexes in Mitochondria. Curr. Med. Chem. 2019, 26, 2918–2932. [Google Scholar] [CrossRef]
  41. Falabella, M.; Kolesar, J.E.; Wallace, C.; de Jesus, D.; Sun, L.; Taguchi, Y.V.; Wang, C.; Wang, T.; Xiang, I.M.; Alder, J.K.; et al. G-Quadruplex Dynamics Contribute to Regulation of Mitochondrial Gene Expression. Sci. Rep. 2019, 9, 5605. [Google Scholar] [CrossRef]
  42. Cui, L.; Zhao, J.; Liu, J. Pyrvinium Sensitizes Clear Cell Renal Cell Carcinoma Response to Chemotherapy Via Casein Kinase 1α-Dependent Inhibition of Wnt/β-Catenin. Am. J. Med. Sci. 2018, 355, 274–280. [Google Scholar] [CrossRef] [PubMed]
  43. Shen, C.; Li, B.; Astudillo, L.; Deutscher, M.P.; Cobb, M.H.; Capobianco, A.J.; Lee, E.; Robbins, D.J. The CK1α Activator Pyrvinium Enhances the Catalytic Efficiency (kcat/Km) of CK1α. Biochemistry 2019, 58, 5102–5106. [Google Scholar] [CrossRef] [PubMed]
  44. Li, J.; Jiang, Y.; Wang, P.; Ke, S.; Yang, L.; Shen, Y. Casein Kinase 1α-Dependent Inhibition of Wnt/β-Catenin Selectively Targets Nasopharyngeal Carcinoma and Increases Chemosensitivity. Anticancer Drugs 2019, 30, e0747. [Google Scholar] [CrossRef] [PubMed]
  45. Lal, S.; Burkhart, R.A.; Beeharry, N.; Bhattacharjee, V.; Londin, E.R.; Cozzitorto, J.A.; Romeo, C.; Jimbo, M.; Norris, Z.A.; Yeo, C.J.; et al. HuR Posttranscriptionally Regulates WEE1: Implications for the DNA Damage Response in Pancreatic Cancer Cells. Cancer Res. 2014, 74, 1128–1140. [Google Scholar] [CrossRef] [PubMed]
  46. Blanco, F.F.; Jimbo, M.; Wulfkuhle, J.; Gallagher, I.; Deng, J.; Enyenihi, L.; Meisner-Kober, N.; Londin, E.; Rigoutsos, I.; Sawicki, J.A.; et al. The mRNA-Binding Protein HuR Promotes Hypoxia-Induced Chemoresistance through Posttranscriptional Regulation of the Proto-Oncogene PIM1 in Pancreatic Cancer Cells. Oncogene 2016, 35, 2529–2541. [Google Scholar] [CrossRef]
  47. Kurosu, T.; Ohga, N.; Hida, Y.; Maishi, N.; Akiyama, K.; Kakuguchi, W.; Kuroshima, T.; Kondo, M.; Akino, T.; Totsuka, Y.; et al. HuR Keeps an Angiogenic Switch on by Stabilising mRNA of VEGF and COX-2 in Tumour Endothelium. Br. J. Cancer 2011, 104, 819–829. [Google Scholar] [CrossRef]
  48. Jones, J.O.; Bolton, E.C.; Huang, Y.; Feau, C.; Guy, R.K.; Yamamoto, K.R.; Hann, B.; Diamond, M.I. Non-Competitive Androgen Receptor Inhibition in Vitro and in Vivo. Proc. Natl. Acad. Sci. USA 2009, 106, 7233–7238. [Google Scholar] [CrossRef]
  49. Pal, S.K.; Tew, B.Y.; Lim, M.; Stankavich, B.; He, M.; Pufall, M.; Hu, W.; Chen, Y.; Jones, J.O. Mechanistic Investigation of the Androgen Receptor DNA-Binding Domain Inhibitor Pyrvinium. ACS Omega 2019, 4, 2472–2481. [Google Scholar] [CrossRef]
  50. Lim, M.; Otto-Duessel, M.; He, M.; Su, L.; Nguyen, D.; Chin, E.; Alliston, T.; Jones, J.O. Ligand-Independent and Tissue-Selective Androgen Receptor Inhibition by Pyrvinium. ACS Chem. Biol. 2014, 9, 692–702. [Google Scholar] [CrossRef] [PubMed]
  51. Zhou, H.-J.; Ren, T.-B. Recent Progress of Cyanine Fluorophores for NIR-II Sensing and Imaging. Chem. Asian J. 2022, 17, e202200147. [Google Scholar] [CrossRef]
  52. Bilici, K.; Cetin, S.; Celikbas, E.; Yagci Acar, H.; Kolemen, S. Recent Advances in Cyanine-Based Phototherapy Agents. Front. Chem. 2021, 9, 707876. [Google Scholar] [CrossRef] [PubMed]
  53. Sulistina, D.R.; Martini, S. The Effect of Rhodamine B on the Cerebellum and Brainstem Tissue of Rattus Norvegicus. J. Public Health Res. 2020, 9, 1812. [Google Scholar] [CrossRef] [PubMed]
  54. Sharma, J.; Sharma, S.; Bhatt, U.; Soni, V. Toxic Effects of Rhodamine B on Antioxidant System and Photosynthesis of Hydrilla Verticillata. J. Hazard. Mater. Lett. 2022, 3, 100069. [Google Scholar] [CrossRef]
  55. Lampidis, T.J.; Bernal, S.D.; Summerhayes, I.C.; Chen, L.B. Selective Toxicity of Rhodamine 123 in Carcinoma Cells in Vitro. Cancer Res. 1983, 43, 716–720. [Google Scholar] [PubMed]
  56. Lampidis, T.J.; Hasin, Y.; Weiss, M.J.; Chen, L.B. Selective Killing of Carcinoma Cells “in Vitro” by Lipophilic-Cationic Compounds: A Cellular Basis. Biomed. Pharmacother. 1985, 39, 220–226. [Google Scholar] [PubMed]
  57. Ayuyan, A.G.; Cohen, F.S. Lipid Peroxides Promote Large Rafts: Effects of Excitation of Probes in Fluorescence Microscopy and Electrochemical Reactions during Vesicle Formation. Biophys. J. 2006, 91, 2172–2183. [Google Scholar] [CrossRef]
  58. Faizi, H.A.; Tsui, A.; Dimova, R.; Vlahovska, P.M. Bending Rigidity, Capacitance, and Shear Viscosity of Giant Vesicle Membranes Prepared by Spontaneous Swelling, Electroformation, Gel-Assisted, and Phase Transfer Methods: A Comparative Study. Langmuir 2022, 38, 10548–10557. [Google Scholar] [CrossRef]
  59. Dimova, R. Giant Vesicles and Their Use in Assays for Assessing Membrane Phase State, Curvature, Mechanics, and Electrical Properties. Annu. Rev. Biophys. 2019, 48, 93–119. [Google Scholar] [CrossRef]
  60. Smith McWilliams, A.D.; Ergülen, S.; Ogle, M.M.; de los Reyes, C.A.; Pasquali, M.; Martí, A.A. Fluorescent Surfactants from Common Dyes—Rhodamine B and Eosin Y. J. Macromol. Sci. Part A Pure Appl. Chem. 2020, 92, 265–274. [Google Scholar] [CrossRef]
  61. Schmeller, T.; Latz-Brüning, B.; Wink, M. Biochemical Activities of Berberine, Palmatine and Sanguinarine Mediating Chemical Defence against Microorganisms and Herbivores. Phytochemistry 1997, 44, 257–266. [Google Scholar] [CrossRef]
  62. Long, J.; Song, J.; Zhong, L.; Liao, Y.; Liu, L.; Li, X. Palmatine: A Review of Its Pharmacology, Toxicity and Pharmacokinetics. Biochimie 2019, 162, 176–184. [Google Scholar] [CrossRef]
  63. Singh, N.; Sharma, B. Toxicological Effects of Berberine and Sanguinarine. Front. Mol. Biosci. 2018, 5, 21. [Google Scholar] [CrossRef]
  64. Chen, X.; Zhang, Y.; Zhu, Z.; Liu, H.; Guo, H.; Xiong, C.; Xie, K.; Zhang, X.; Su, S. Protective Effect of Berberine on Doxorubicin-induced Acute Hepatorenal Toxicity in Rats. Mol. Med. Rep. 2016, 13, 3953–3960. [Google Scholar] [CrossRef] [PubMed]
  65. Hasanein, P.; Ghafari-Vahed, M.; Khodadadi, I. Effects of Isoquinoline Alkaloid Berberine on Lipid Peroxidation, Antioxidant Defense System, and Liver Damage Induced by Lead Acetate in Rats. Redox Rep. 2017, 22, 42–50. [Google Scholar] [CrossRef] [PubMed]
  66. Verma, S.K.; Dev, G.; Tyagi, A.K.; Goomber, S.; Jain, G.V. Argemone Mexicana Poisoning: Autopsy Findings of Two Cases. Forensic Sci. Int. 2001, 115, 135–141. [Google Scholar] [CrossRef]
  67. Wang, J.; He, H.; Xiang, C.; Fan, X.-Y.; Yang, L.-Y.; Yuan, L.; Jiang, F.-L.; Liu, Y. Uncoupling Effect of F16 Is Responsible for Its Mitochondrial Toxicity and Anticancer Activity. Toxicol. Sci. 2018, 161, 431–442. [Google Scholar] [CrossRef] [PubMed]
  68. Dubinin, M.V.; Nedopekina, D.A.; Ilzorkina, A.I.; Semenova, A.A.; Sharapov, V.A.; Davletshin, E.V.; Mikina, N.V.; Belsky, Y.P.; Spivak, A.Y.; Akatov, V.S.; et al. Conjugation of Triterpenic Acids of Ursane and Oleanane Types with Mitochondria-Targeting Cation F16 Synergistically Enhanced Their Cytotoxicity against Tumor Cells. Membranes 2023, 13, 563. [Google Scholar] [CrossRef] [PubMed]
  69. Chen, H.; Wang, J.; Feng, X.; Zhu, M.; Hoffmann, S.; Hsu, A.; Qian, K.; Huang, D.; Zhao, F.; Liu, W.; et al. Mitochondria-Targeting Fluorescent Molecules for High Efficiency Cancer Growth Inhibition and Imaging. Chem. Sci. 2019, 10, 7946–7951. [Google Scholar] [CrossRef]
  70. Gureev, A.P.; Shaforostova, E.A.; Laver, D.A.; Khorolskaya, V.G.; Syromyatnikov, M.Y.; Popov, V.N. Methylene Blue Elicits Non-Genotoxic H2O2 Production and Protects Brain Mitochondria from Rotenone Toxicity. J. Appl. Biomed. 2019, 17, 107–114. [Google Scholar] [CrossRef]
  71. Gureev, A.P.; Sadovnikova, I.S.; Popov, V.N. Molecular Mechanisms of the Neuroprotective Effect of Methylene Blue. Biochemistry 2022, 87, 940–956. [Google Scholar] [CrossRef]
  72. Sanchala, D.; Bhatt, L.K.; Pethe, P.; Shelat, R.; Kulkarni, Y.A. Anticancer Activity of Methylene Blue via Inhibition of Heat Shock Protein 70. Biomed. Pharmacother. 2018, 107, 1037–1045. [Google Scholar] [CrossRef] [PubMed]
  73. Jang, D.H.; Nelson, L.S.; Hoffman, R.S. Methylene Blue for Distributive Shock: A Potential New Use of an Old Antidote. J. Med. Toxicol. 2013, 9, 242–249. [Google Scholar] [CrossRef]
  74. Martins, W.K.; Santos, N.F.; Rocha, C.d.S.; Bacellar, I.O.L.; Tsubone, T.M.; Viotto, A.C.; Matsukuma, A.Y.; Abrantes, A.B.d.P.; Siani, P.; Dias, L.G.; et al. Parallel Damage in Mitochondria and Lysosomes Is an Efficient Way to Photoinduce Cell Death. Autophagy 2019, 15, 259–279. [Google Scholar] [CrossRef] [PubMed]
  75. West, T.J.; Bi, J.; Martínez-Peña, F.; Curtis, E.J.; Gazaniga, N.R.; Mischel, P.S.; Lairson, L.L. A Cell Type Selective YM155 Prodrug Targets Receptor-Interacting Protein Kinase 2 to Induce Brain Cancer Cell Death. J. Am. Chem. Soc. 2023, 145, 8355–8363. [Google Scholar] [CrossRef] [PubMed]
  76. Mondal, A.; Jia, D.; Bhatt, V.; Akel, M.; Roberge, J.; Guo, J.Y.; Langenfeld, J. Ym155 Localizes to the Mitochondria Leading to Mitochondria Dysfunction and Activation of AMPK That Inhibits BMP Signaling in Lung Cancer Cells. Sci. Rep. 2022, 12, 13135. [Google Scholar] [CrossRef] [PubMed]
  77. Wani, T.H.; Chowdhury, G.; Chakrabarty, A. Generation of Reactive Oxygen Species Is the Primary Mode of Action and Cause of Survivin Suppression by Sepantronium Bromide (YM155). RSC Med. Chem. 2021, 12, 566–578. [Google Scholar] [CrossRef] [PubMed]
  78. Sharma, S.; Zhang, Y.; Akter, K.A.; Nozohouri, S.; Archie, S.R.; Patel, D.; Villalba, H.; Abbruscato, T. Permeability of Metformin across an In Vitro Blood-Brain Barrier Model during Normoxia and Oxygen-Glucose Deprivation Conditions: Role of Organic Cation Transporters (Octs). Pharmaceutics 2023, 15, 1357. [Google Scholar] [CrossRef] [PubMed]
  79. Feng, J.; Wang, X.; Ye, X.; Ares, I.; Lopez-Torres, B.; Martínez, M.; Martínez-Larrañaga, M.-R.; Wang, X.; Anadón, A.; Martínez, M.-A. Mitochondria as an Important Target of Metformin: The Mechanism of Action, Toxic and Side Effects, and New Therapeutic Applications. Pharmacol. Res. 2022, 177, 106114. [Google Scholar] [CrossRef]
  80. Zhao, B.; Luo, J.; Yu, T.; Zhou, L.; Lv, H.; Shang, P. Anticancer Mechanisms of Metformin: A Review of the Current Evidence. Life Sci. 2020, 254, 117717. [Google Scholar] [CrossRef]
  81. Teng, X.; Brown, J.; Morel, L. Redox Homeostasis Involvement in the Pharmacological Effects of Metformin in Systemic Lupus Erythematosus. Antioxid. Redox Signal. 2022, 36, 462–479. [Google Scholar] [CrossRef]
  82. Silachev, D.N.; Khailova, L.S.; Babenko, V.A.; Gulyaev, M.V.; Kovalchuk, S.I.; Zorova, L.D.; Plotnikov, E.Y.; Antonenko, Y.N.; Zorov, D.B. Neuroprotective Effect of Glutamate-Substituted Analog of Gramicidin A Is Mediated by the Uncoupling of Mitochondria. Biochim. Biophys. Acta 2014, 1840, 3434–3442. [Google Scholar] [CrossRef]
  83. Khailova, L.S.; Rokitskaya, T.I.; Kovalchuk, S.I.; Kotova, Е.А.; Sorochkina, A.I.; Antonenko, Y.N. Role of Mitochondrial Outer Membrane in the Uncoupling Activity of N-Terminally Glutamate-Substituted Gramicidin A. Biochim. Biophys. Acta Biomembr. 2019, 1861, 281–287. [Google Scholar] [CrossRef]
  84. Zhang, C.-S.; Li, M.; Ma, T.; Zong, Y.; Cui, J.; Feng, J.-W.; Wu, Y.-Q.; Lin, S.-Y.; Lin, S.-C. Metformin Activates AMPK through the Lysosomal Pathway. Cell Metab. 2016, 24, 521–522. [Google Scholar] [CrossRef]
  85. Patel, S.; Singh, N.; Kumar, L. Evaluation of Effects of Metformin in Primary Ovarian Cancer Cells. Asian Pac. J. Cancer Prev. 2015, 16, 6973–6979. [Google Scholar] [CrossRef] [PubMed]
  86. Xiao, Y.; Liu, F.; Kong, Q.; Zhu, X.; Wang, H.; Li, S.; Jiang, N.; Yu, C.; Yun, L. Metformin Induces S-Adenosylmethionine Restriction to Extend the Caenorhabditis Elegans Healthspan through H3K4me3 Modifiers. Aging Cell 2022, 21, e13567. [Google Scholar] [CrossRef] [PubMed]
  87. Alves, A.C.; Magarkar, A.; Horta, M.; Lima, J.L.F.C.; Bunker, A.; Nunes, C.; Reis, S. Influence of Doxorubicin on Model Cell Membrane Properties: Insights from in Vitro and in Silico Studies. Sci. Rep. 2017, 7, 6343. [Google Scholar] [CrossRef] [PubMed]
  88. Han, M.; Vakili, M.R.; Soleymani Abyaneh, H.; Molavi, O.; Lai, R.; Lavasanifar, A. Mitochondrial Delivery of Doxorubicin via Triphenylphosphine Modification for Overcoming Drug Resistance in MDA-MB-435/DOX Cells. Mol. Pharm. 2014, 11, 2640–2649. [Google Scholar] [CrossRef]
  89. Gaytan, S.L.; Lawan, A.; Chang, J.; Nurunnabi, M.; Bajpeyi, S.; Boyle, J.B.; Han, S.M.; Min, K. The Beneficial Role of Exercise in Preventing Doxorubicin-Induced Cardiotoxicity. Front. Physiol. 2023, 14, 1133423. [Google Scholar] [CrossRef]
  90. Wang, X.; Tian, X.; Wu, Y.; Shen, X.; Yang, S.; Chen, S. Enhanced Doxorubicin Production by Streptomyces Peucetius Using a Combination of Classical Strain Mutation and Medium Optimization. Prep. Biochem. Biotechnol. 2018, 48, 514–521. [Google Scholar] [CrossRef]
  91. Sohail, M.; Sun, Z.; Li, Y.; Gu, X.; Xu, H. Research Progress in Strategies to Improve the Efficacy and Safety of Doxorubicin for Cancer Chemotherapy. Expert Rev. Anticancer Ther. 2021, 21, 1385–1398. [Google Scholar] [CrossRef]
  92. Johnson-Arbor, K.; Dubey, R. Doxorubicin; StatPearls Publishing: St. Petersburg, FL, USA, 2023. [Google Scholar]
  93. Liu, H.-N.; Guo, N.-N.; Wang, T.-T.; Guo, W.-W.; Lin, M.-T.; Huang-Fu, M.-Y.; Vakili, M.R.; Xu, W.-H.; Chen, J.-J.; Wei, Q.-C.; et al. Mitochondrial Targeted Doxorubicin-Triphenylphosphonium Delivered by Hyaluronic Acid Modified and pH Responsive Nanocarriers to Breast Tumor: In Vitro and in Vivo Studies. Mol. Pharm. 2018, 15, 882–891. [Google Scholar] [CrossRef]
  94. Arafa, K.K.; Smyth, H.D.C.; El-Sherbiny, I.M. Mitotropic Triphenylphosphonium Doxorubicin-Loaded Core-Shell Nanoparticles for Cellular and Mitochondrial Sequential Targeting of Breast Cancer. Int. J. Pharm. 2021, 606, 120936. [Google Scholar] [CrossRef] [PubMed]
  95. Peng, X.; Tang, S.; Tang, D.; Zhou, D.; Li, Y.; Chen, Q.; Wan, F.; Lukas, H.; Han, H.; Zhang, X.; et al. Autonomous Metal-Organic Framework Nanorobots for Active Mitochondria-Targeted Cancer Therapy. Sci. Adv. 2023, 9, eadh1736. [Google Scholar] [CrossRef]
  96. Priya, L.B.; Baskaran, R.; Huang, C.-Y.; Padma, V.V. Neferine Ameliorates Cardiomyoblast Apoptosis Induced by Doxorubicin: Possible Role in Modulating NADPH oxidase/ROS-Mediated NFκB Redox Signaling Cascade. Sci. Rep. 2017, 7, 12283. [Google Scholar] [CrossRef]
  97. Montalvo, R.N.; Doerr, V.; Min, K.; Szeto, H.H.; Smuder, A.J. Doxorubicin-Induced Oxidative Stress Differentially Regulates Proteolytic Signaling in Cardiac and Skeletal Muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2020, 318, R227–R233. [Google Scholar] [CrossRef]
  98. Jeon, K.-H.; Yu, H.V.; Kwon, Y. Hyperactivated M-Calpain Affects Acquisition of Doxorubicin Resistance in Breast Cancer Cells. Biochim. Biophys. Acta Gen. Subj. 2018, 1862, 1126–1133. [Google Scholar] [CrossRef] [PubMed]
  99. Wei, Y.; Zhao, J.; Xiong, J.; Chai, J.; Yang, X.; Wang, J.; Chen, J.; Wang, J. Wogonin Reduces Cardiomyocyte Apoptosis from Mitochondrial Release of Cytochrome c to Improve Doxorubicin-Induced Cardiotoxicity. Exp. Ther. Med. 2022, 23, 205. [Google Scholar] [CrossRef] [PubMed]
  100. Ye, X.; Li, Y.; Lv, B.; Qiu, B.; Zhang, S.; Peng, H.; Kong, W.; Tang, C.; Huang, Y.; Du, J.; et al. Endogenous Hydrogen Sulfide Persulfidates Caspase-3 at Cysteine 163 to Inhibit Doxorubicin-Induced Cardiomyocyte Apoptosis. Oxid. Med. Cell. Longev. 2022, 2022, 6153772. [Google Scholar] [CrossRef]
  101. Nishida, K.; Kyoi, S.; Yamaguchi, O.; Sadoshima, J.; Otsu, K. The Role of Autophagy in the Heart. Cell Death Differ. 2009, 16, 31–38. [Google Scholar] [CrossRef]
  102. Chen, L.B. Mitochondrial Membrane Potential in Living Cells. Annu. Rev. Cell Biol. 1988, 4, 155–181. [Google Scholar] [CrossRef]
  103. Sun, X.; Wong, J.R.; Song, K.; Hu, J.; Garlid, K.D.; Chen, L.B. AA1, a Newly Synthesized Monovalent Lipophilic Cation, Expresses Potent in Vivo Antitumor Activity. Cancer Res. 1994, 54, 1465–1471. [Google Scholar]
  104. Modica-Napolitano, J.S.; Koya, K.; Weisberg, E.; Brunelli, B.T.; Li, Y.; Chen, L.B. Selective Damage to Carcinoma Mitochondria by the Rhodacyanine MKT-077. Cancer Res. 1996, 56, 544–550. [Google Scholar]
  105. Weiss, M.J.; Wong, J.R.; Ha, C.S.; Bleday, R.; Salem, R.R.; Steele, G.D., Jr.; Chen, L.B. Dequalinium, a Topical Antimicrobial Agent, Displays Anticarcinoma Activity Based on Selective Mitochondrial Accumulation. Proc. Natl. Acad. Sci. USA 1987, 84, 5444–5448. [Google Scholar] [CrossRef] [PubMed]
  106. Fantin, V.R.; Berardi, M.J.; Scorrano, L.; Korsmeyer, S.J.; Leder, P. A Novel Mitochondriotoxic Small Molecule That Selectively Inhibits Tumor Cell Growth. Cancer Cell 2002, 2, 29–42. [Google Scholar] [CrossRef]
  107. Kollmitzer, B.; Heftberger, P.; Rappolt, M.; Pabst, G. Monolayer Spontaneous Curvature of Raft-Forming Membrane Lipids. Soft Matter 2013, 9, 10877–10884. [Google Scholar] [CrossRef]
  108. Kaltenegger, M.; Kremser, J.; Frewein, M.P.K.; Ziherl, P.; Bonthuis, D.J.; Pabst, G. Intrinsic Lipid Curvatures of Mammalian Plasma Membrane Outer Leaflet Lipids and Ceramides. Biochim. Biophys. Acta Biomembr. 2021, 1863, 183709. [Google Scholar] [CrossRef] [PubMed]
  109. Zhelev, D.V.; Needham, D. Tension-Stabilized Pores in Giant Vesicles: Determination of Pore Size and Pore Line Tension. Biochim. Biophys. Acta 1993, 1147, 89–104. [Google Scholar] [CrossRef]
  110. Rice, A.; Zimmerberg, J.; Pastor, R.W. Initiation and Evolution of Pores Formed by Influenza Fusion Peptides Probed by Lysolipid Inclusion. Biophys. J. 2023, 122, 1018–1032. [Google Scholar] [CrossRef]
  111. Jiménez-Munguía, I.; Volynsky, P.E.; Batishchev, O.V.; Akimov, S.A.; Korshunova, G.A.; Smirnova, E.A.; Knorre, D.A.; Sokolov, S.S.; Severin, F.F. Effects of Sterols on the Interaction of SDS, Benzalkonium Chloride, and A Novel Compound, Kor105, with Membranes. Biomolecules 2019, 9, 627. [Google Scholar] [CrossRef] [PubMed]
  112. Dupont, C.-H.; Mazat, J.P.; Guerin, B. The Role of Adenine Nucleotide Translocation in the Energization of the Inner Membrane of Mitochondria Isolated from ϱ+ and ϱo Strains of Saccharomyces Cerevisiae. Biochem. Biophys. Res. Commun. 1985, 132, 1116–1123. [Google Scholar] [CrossRef]
  113. Shrestha, R.; Johnson, E.; Byrne, F.L. Exploring the Therapeutic Potential of Mitochondrial Uncouplers in Cancer. Mol. Metab. 2021, 51, 101222. [Google Scholar] [CrossRef] [PubMed]
  114. Knorre, D.A.; Besedina, E.; Karavaeva, I.E.; Smirnova, E.A.; Markova, O.V.; Severin, F.F. Alkylrhodamines Enhance the Toxicity of Clotrimazole and Benzalkonium Chloride by Interfering with Yeast Pleiotropic ABC-Transporters. FEMS Yeast Res. 2016, 16, fow030. [Google Scholar] [CrossRef] [PubMed]
  115. Sukjoi, W.; Ngamkham, J.; Attwood, P.V.; Jitrapakdee, S. Targeting Cancer Metabolism and Current Anti-Cancer Drugs. Adv. Exp. Med. Biol. 2021, 1286, 15–48. [Google Scholar] [PubMed]
  116. Szarka, A.; Kapuy, O.; Lőrincz, T.; Bánhegyi, G. Vitamin C and Cell Death. Antioxid. Redox Signal. 2021, 34, 831–844. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Сhemical structures of lipocations.
Figure 1. Сhemical structures of lipocations.
Membranes 13 00841 g001
Table 1. Toxic effects of the most well-studied penetrating cations. See text for details.
Table 1. Toxic effects of the most well-studied penetrating cations. See text for details.
Triphenyl phosphonium Dequalinium chloride Pyrvinium Rhodamine Berberine F16 Methylene blue YM155 Doxorubicin Metformin
Proton conductivity
Inhibition of respiratory Chain
DNA damage
Inhibition of MDR pumps
Inhibition of soluble (non-membrane bound) proteins
Disruption of phospholipid bilayers
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

Sokolov, S.; Zyrina, A.; Akimov, S.; Knorre, D.; Severin, F. Toxic Effects of Penetrating Cations. Membranes 2023, 13, 841. https://doi.org/10.3390/membranes13100841

AMA Style

Sokolov S, Zyrina A, Akimov S, Knorre D, Severin F. Toxic Effects of Penetrating Cations. Membranes. 2023; 13(10):841. https://doi.org/10.3390/membranes13100841

Chicago/Turabian Style

Sokolov, Svyatoslav, Anna Zyrina, Sergey Akimov, Dmitry Knorre, and Fedor Severin. 2023. "Toxic Effects of Penetrating Cations" Membranes 13, no. 10: 841. https://doi.org/10.3390/membranes13100841

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