Next Article in Journal
Simultaneous Determination of Fucoxanthin and Its Deacetylated Metabolite Fucoxanthinol in Rat Plasma by Liquid Chromatography-Tandem Mass Spectrometry
Previous Article in Journal
Alkaloids with Cardiovascular Effects from the Marine-Derived Fungus Penicillium expansum Y32
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell Death Inducing Microbial Protein Phosphatase Inhibitors—Mechanisms of Action

1
Department of Biomedicine, University of Bergen, Jonas Lies vei 81, N-5009 Bergen, Norway
2
Department of Clinical Science, University of Bergen, Jonas Lies vei 65, N-5021 Bergen, Norway
*
Author to whom correspondence should be addressed.
Mar. Drugs 2015, 13(10), 6505-6520; https://doi.org/10.3390/md13106505
Submission received: 11 September 2015 / Revised: 12 October 2015 / Accepted: 15 October 2015 / Published: 22 October 2015

Abstract

:
Okadaic acid (OA) and microcystin (MC) as well as several other microbial toxins like nodularin and calyculinA are known as tumor promoters as well as inducers of apoptotic cell death. Their intracellular targets are the major serine/threonine protein phosphatases. This review summarizes mechanisms believed to be responsible for the death induction and tumor promotion with focus on the interdependent production of reactive oxygen species (ROS) and activation of Ca2+/calmodulin kinase II (CaM-KII). New data are presented using inhibitors of specific ROS producing enzymes to curb nodularin/MC-induced liver cell (hepatocyte) death. They indicate that enzymes of the arachidonic acid pathway, notably phospholipase A2, 5-lipoxygenase, and cyclooxygenases, may be required for nodularin/MC-induced (and presumably OA-induced) cell death, suggesting new ways to overcome at least some aspects of OA and MC toxicity.

Graphical Abstract

1. Introduction

The dinoflagellate toxin okadaic acid (OA) and the toxic cyanobacterial microcystins (MC, including nodularin), pose a health hazard to both humans and livestock [1]. Although OA and MC/Nod are produced by organisms from two different kingdoms and have disparate primary structure, their main target is the same; phospho-Ser/-Thr protein phosphatases (PPs), foremost the PP1s and PP2As of this PP class. The targeted PPs are pivotal in eukaryote signaling, where they are controlled by hundreds of distinct regulatory scaffolding subunits as well as endogenous inhibitory peptides [2,3].

2. Structural Features of Microbial Toxins

OA and MC are relatively complex structures (Figure 1A,B), with a molecular weight of about 800 and 1000 Da, respectively. OA is a polyether with a terminal carboxy-moiety [4]. MC is a family of cyclic heptapeptides [5,6] with a general structure of cyclo-(d-Ala1-Xaa2-d-MeAsp3-Yaa4-Adda5-d-Glu6-Mdha7) where the amino acids in the two and four position are variable. The most common structure is MC-LeuArg (MC-LR), but other variants such as TyrArg, LeuTrp, and LeuAla are also common. MC contains several non-proteinogenic amino acids, such as Adda (3-amino-9-methoxy-2,6,8-trimethyl-10-phenyl-4,6-decadienoic acid), d-MeAsp (d-erythro-β-methylaspartic acid), and Mdha (N-methyldehydroalanine). The related PP inhibiting cyanobacterial cyclic pentapeptide nodularin (Nod) shares several non-proteinogenic amino acids with MC, such as Adda and d-MeAsp, but instead of Mdha, the dehydro-amino acid Mdhb (2-(methylamino)-2-dehydrobutyric acid) [7,8,9].
The striking similar action of these distinct compounds, which differ in primary structure, becomes understandable upon inspection of their in solution structure, and their docking into the PP active site (Figure 1A,B). The structures of the two toxins in complex with PP1 reveal similar three-dimensional conformation (Figure 1C,D). Either compound has important interactions with the hydrophobic groove and the β12β13-loop of PP, and covers the catalytic site (for details of the toxin binding to PP1 or PP2A see: [10,11,12,13,14]). An interesting feature of MC is the ability of its methyl-dehydroalanine moiety to form a covalent linkage with Cys273 on PP1, leading to irreversible phosphatase inhibition [15]. Nodularin has a similar inhibitory profile as microcystin [16], but the replacement of the N-methyldehydroalanine by N-methyldehydrobutyrine prevents the formation of a covalent linkage with Cys273.

3. Comparative Activity of OA and MC-LR against Various Protein Phosphatases

OA and MC-LR have been compared for their ability to inhibit a number of PPs (see Table 1 for reported IC50 values). MC-LR has high potency (IC50 ≤ 1 nM) against PP1, PP2A, PP3, PP4 and PP5, and may therefore be the inhibitor of choice to obtain a more generalized PP inhibition (note, however, that neither OA nor MC show significant inhibition of PP2B/calcineurin, PP7, or PP2C (Table 1). OA shows about 100-fold preference for PP2A and PP4 compared to PP1 (Table 1). Effects obtained at low OA concentrations are therefore unlikely to be caused by PP1 inhibition.
Figure 1. (A,B) Structure of okadaic acid (A) and microcystin (B); (C,D) the three-dimensional representation of okadaic acid (C) and microcystin (D) when complexed with protein phosphatase 1 (PP1). The OA-PP1structure is from [14], and the MC-LR-PP1 structure is from [13].
Figure 1. (A,B) Structure of okadaic acid (A) and microcystin (B); (C,D) the three-dimensional representation of okadaic acid (C) and microcystin (D) when complexed with protein phosphatase 1 (PP1). The OA-PP1structure is from [14], and the MC-LR-PP1 structure is from [13].
Marinedrugs 13 06505 g001
Table 1. Reported IC50 values (nM) of okadaic acid and microcystin LR for inhibition of various serine-threonine PP family members.
Table 1. Reported IC50 values (nM) of okadaic acid and microcystin LR for inhibition of various serine-threonine PP family members.
PP family Member 1IC50 (nM)
Okadaic acidReferencesMC-LRReferences
PP115–50[17,18]0.3–2[15,17,18,19,20]
PP2A0.1–0.3[17,18]0.05–1[17,18,19,20]
PP2B~4000[17]~2000[17,19]
PP33.7–4[18]0.2[18]
PP40.1[17,21]0.15[17,21]
PP53.5[17]1.0[17]
PP7>1000[17,22]>1000[17,22]
1 Data for PP2C (a member of the PPM family of phosho-protein phosphatases) have, to the best of our knowledge, not been published, but one report states that nodularin lacks activity towards this phosphatase [16].
Most inhibition assays require a substantial concentration of phosphatase (typically near 1 nM) to achieve significant dephosphorylation. This may lead to under-estimation of the IC50 of unbound toxin (see [23] for details). Data obtained by a competition assay between various toxins and [125I]-labeled MC-YR at very low PP2A concentrations (Table 2) illustrate this point. This assay reveals that MC-LR binds with even higher affinity (IC50 = 0.017 nM) to PP2A (Table 2) than revealed by inhibition assays (Table 1). Note also that nodularin is a high affinity PP2A inhibitor (IC50 = 0.015 nM) and that tautomycin, often considered a preferential PP1 inhibitor [19], has IC50 < 0.6 nM for PP2A (Table 2).
Table 2. IC50 values for various phosphatase inhibitors for binding to PP2A.
Table 2. IC50 values for various phosphatase inhibitors for binding to PP2A.
CompoundIC50 (nM)
Nodularin0.015
MC-LR0.017
MC-YR0.075
OA0.100
CalyculinA0.250
Tautomycin0.560
Data are from [23], and are based on competitive displacement of [125I]-labeled MC-YR from purified PP2A.

4. Cellular Uptake of Microbial Phosphatase Inhibitors

The mechanisms of how OA and MC toxins enter cells are fundamentally different. Okadaic acid is amphiphilic [24] enough to cross membranes. Membrane interactions of MC have been reported [25], likely through the insertion of the hydrophobic Adda moiety. MC lacks, however, the ability of the lipopeptide anabaenolysin to permeabilize membranes in a cholesterol-dependent manner [26]. In fact, MC requires transporter proteins to enter cells [27,28]. This may be due to both its charge and polarity and its bulk, since addition of a hydrophobic acyloxy-methyl ester on the carboxyl-groups or phenyl-glyoxal on the arginine of MC or nodularin did not improve significantly the entry into cells lacking MC transporters [29]. As a consequence of their different membrane permeability OA can induce cell death with apoptotic features [30] in virtually all cell types, whereas MC only induces apoptosis in cells expressing the liver specific organic anion transporter polypetides (OATP) 1B1 or 1B3 [28]. For cell experiments, this means that only primary hepatocytes, or cells with enforced expression of the transporters are sensitive to MC at nM concentrations, while mammalian cells not expressing OATP1B1/3 require concentrations above 10 μM [31,32,33]. For this reason isolated primary hepatocytes have been used as an alternative to mouse survival test to detect MC/nodularin or diarrhetic shellfish poisons like OA, using the rapid induction of apoptosis as read-out [34].
MC and nodularin depend on hepatocyte OATP1B1/3 for cell entry [27] OATP1B1/3 inhibitors will protect hepatocytes against both nodularin and MC induced apoptosis [35]. It is of interest that some cyanobacterial nostocyclopeptides are OATP1B1/3 inhibitors whose original function may have been to block nutrient channels of their primitive eukaryotic predators [36]. One may consider MC and nodularin as competitive channel blocking toxins that have acquired the additional property of inhibiting the major PPs in predator cells. The extreme conservation of the active site of PP2A and PP1 through evolution explains why these ancient toxins also act on human phosphatases.

5. Features of Microbial PP Inhibitor-Induced Cell Death

As first reported in [30] the phosphatase inhibitors OA and MC-LR induce cellular apoptosis. The apoptosis was independent of new protein synthesis and therefore not mediated by the execution of a genetic program. The OA-or MC/nodularin-induced death had classical morphological features of apoptosis such as cell and nuclear shrinkage, chromatin condensation, formation and shedding of apoptotic bodies, annexin-V externalization, but lacked DNA fragmentation [30,37,38]. Internucleosomal DNA fragmentation and 28S ribosomal RNA cleavage was observed in some, but not all, OA-treated leukemia cell lines, suggesting that this trait is cell type specific [39,40].
Secondary necrosis (apo-necrosis) is a late phenomenon in MC or nodularin exposed hepatocytes [38]. In contrast, we have observed that primary hepatocytes exposed to hypoxia during isolation responded to MC/nodularin with primary necrotic death, characterized by membrane leakage and secondary DNA fragmentation. This is reminiscent of the switch from an apoptotic to a necrotic response to tumor necrosis factor α in cells with depleted energy stores or inactive caspase 8 (see [41] for recent review). Inactive caspase 8 cannot alone explain the necrotic phenotype of hypoxic hepatocytes since the caspase inhibitor z-VAD-fmk had little effect on MC-induced hepatocyte apoptosis in normoxic hepatocytes [37]. The features of PP-inhibitor-induced death depend not only on the target cell and its energy state, but also on the concentration and nature of the inhibitor. Thus, while acute myeloid leukemia (AML) cells incubated with high concentration of calyculin A (CalA) lacked the nuclear envelope and had increased ATP content, OA or CalA at low concentration induced DNA and RNA cleavage, nuclear fragmentation, and cell shrinkage [42].

6. The Pathogenesis of the Acute Hepatotoxicity of MC and Nodularin

The ingestion of MC or Nod producing cyanobacteria can cause lethal acute liver damage. The death occurs too rapidly to be explained by hepatocyte metabolic failure alone, and is probably due to disintegration of the liver micro-architecture, accompanied by lethal bleeding [43,44]. As first shown by Eriksson [45], MC causes strong condensation of hepatocyte actomyosin filaments accompanied by blebbing [46]. Cell blebbing was also noted in an early study by Runnegar et al. [47]. The actomyosin contraction may be related to myosin light chain hyperphosphorylation, which coincides with cell shrinkage and blebbing, and is one of the first phosphorylation events detected in hepatocytes exposed to MC [37]. Both shrinkage and blebbing depend on high intracellular pressure, which can be produced by general hypercontraction of the subplasmalemnal actomyosin web.
Loosening of cell-cell and cell-substratum contacts has been observed in hepatocyte monolayers [48]. In fact, hepatocytes exposed for only 30 min to 0.5 µM MC-LR remained irreversibly shrunk and blebbed, and were unable to attach to collagen substratum [38]. The combination of hepatocyte shrinkage and detachment is expected to disrupt bile canaliculi and blood vessels, resulting in lethal leakage of blood and toxic bile acids. The PP-inhibitor responses noted above are not unique for hepatocytes [49].
It is questionable if mitochondrial are early targets in PP-inhibitor induced cell death. Thus, OA or CalA failed to synergize with cAMP to induce cell death in AML cells [42], in which cAMP acts by inducing the expression of the pro-apoptotic Bcl-2 protein Bim, with subsequent mitochondrial damage [50]. Additionally, cells overexpressing the mitochondria-protecting B-cell lymphoma (Bcl)-2 or Bcl-XL were equally sensitive to microinjected nodularin as wt cells [37]. Micro-mitochondriosis, induced by mitochondrial fission, has, however, been observed in AML cells exposed to high concentration of CalA [42] and in neurons exposed to OA [51].

7. PP Inhibitors as Tumor Promoters

OA became early known as a promoter of skin tumors [52], and MC as a potential hepatocarcinoma promoter [53]. The discovery of the mitogen activated protein kinase (MAPK) cascade and its antagonism by PP2A, and that the oncogenic SV40 small T antigen was a PP2A inhibitor [54,55], led to the assumption that OA and MC tumor promotion was mediated mainly by stimulation of MAPK to enhance cell proliferation and survival. The assumption is supported by studies showing that CalA-enhanced ERK activity may protect U937 cells against death [56] and that MC may enhance slightly hepatocyte proliferation [57]. OA can, however, even at sub-apoptotic concentration, inhibit cellular DNA replication [57,58]. The dualistic effects of MC-LR and OA have been reviewed by Gehringer [59]. It is also possible that PP inhibitor induced cell damage leads to compensatory growth of less damaged neighboring cells. Other mechanisms whereby OA can promote tumor formation are by interference with normal centrosome duplication and chromosome segregation during mitosis [60,61].

8. The Role of CaM-KII in PP Inhibitor Action—Is CaM-KII an Amplifier of ROS Generation?

Phosphatase inhibition can increase phosphorylation of a protein only if the relevant protein kinase(s) is active. The ubiquitously expressed Ca2+/calmodulin dependent protein kinase II (CaM-KII) was found essential for OA-, MC- and nodularin- induced apoptosis and early phosphorylation events in a number of cell types, as well as for the OA-induced inhibition of hepatocyte DNA replication [38,57,62]. Since CaM-KII autophosphorylation on Thr286 [63] is reversed by several phosphatases, including PP1 and PP2A [63], it is expected that PP inhibitors activate CaM-KII by enhancing the activating autophosphorylation on Thr286 [63]. More surprising is that CaM-KII inhibitors also can block PP inhibitor induced protein phosphorylation events catalyzed by other protein kinases. One mechanism of CaM-KII action is through direct activating phosphorylation of other kinases such as myosin light chain kinase [38,62], which may explain the early hyperphosphorylation of myosin light chain in MC- or nodularin- exposed cells [38,62]. Another mechanism is through generation of reactive oxygen species (ROS), which also feedback on CaM-KII itself as it is subject to activation by oxidation of Met281/282 [64].
Protein phosphatases are inhibited by ROS that target specific redox sensitive amino acids. The reversible inhibitory oxidation of protein tyrosine phosphatases by hydrogen peroxide was discovered in the mid-1990s, whereas redox regulation of Ser/Thr phosphatases is a more recent discovery [65]. The PP2A regulatory subunit B56δ is reported to become tyrosine nitrated, leading to poor holoenzyme formation and substrate targeting [66]. The direct inhibition of PP2A catalytic activity by hydrogen peroxide has also been reported, leading to the activation of cellular stress signaling pathways [67].
Several cellular activities can produce ROS (Figure 2A) and are, therefore, potential sources of the ROS increase by PP inhibitors. The extent to which locally produced ROS accumulates will depend on the presence of nearby enzymes such as superoxide dismutase, catalase, the selenocysteine containing glutathione peroxidases and thioredoxin reductases, which catalyze the conversion of different ROS types to harmless molecules. In addition, glutathione, N-acetyl-cysteine and redox active proteins like peroxyredoxins and methionine sulfoxide reductases can facilitate the normalization of ROS modified proteins. PP inhibitors can therefore enhance ROS accumulation also by inhibiting the action of ROS scavengers or neutralizers.
Figure 2. ROS-producing enzymes in PPI-induced apoptosis. (A) Illustrates some of the major cellular sources of reactive oxygen species (ROS). The illustration focuses on different ROS producing enzymes; NADPH oxidase (NOX), nitric oxide synthase (NOS), xanthine oxidase (XO), monoamine oxidase (MAO), cyclooxygenases (COX), 12-Lipoxygenase (12LOX), 5-Lipoxygenase (5LOX), and microsomal Cytochrome P450 complexes (CYP). Cellular phospholipase A2 (PLA2) does not generate ROS, but acts upstream of COX and 5LOX. The numbers in bold refers to the enzyme that is targeted by the inhibitors used in panel. (B). The effect of different inhibitors towards ROS producing enzymes was tested against nodularin (Nod, 100 nM, 75 min) induced apoptosis in isolated primary rat hepatocytes. The inhibitors (assigned with numbers) were: (1) Diphenyleneiodonium chloride (25 µM), (2) allopurinol (100 µM), (3) metyrapone (500 µM), (4) baicalein (5 µM), (5) AA861 (5 µM), (6) Indomethacine (10 µM), (7) nordihydroguaiaretic acid (20 µM), (8) arachidonyl trifluoromethylketone (5 µM), and (9) naringin (100 µM). After treatment, the cells were fixed evaluated for apoptosis and necrosis by microscopy. Treatment with nodularin gave rise to 86% apoptosis, compared to less than 1% in control.
Figure 2. ROS-producing enzymes in PPI-induced apoptosis. (A) Illustrates some of the major cellular sources of reactive oxygen species (ROS). The illustration focuses on different ROS producing enzymes; NADPH oxidase (NOX), nitric oxide synthase (NOS), xanthine oxidase (XO), monoamine oxidase (MAO), cyclooxygenases (COX), 12-Lipoxygenase (12LOX), 5-Lipoxygenase (5LOX), and microsomal Cytochrome P450 complexes (CYP). Cellular phospholipase A2 (PLA2) does not generate ROS, but acts upstream of COX and 5LOX. The numbers in bold refers to the enzyme that is targeted by the inhibitors used in panel. (B). The effect of different inhibitors towards ROS producing enzymes was tested against nodularin (Nod, 100 nM, 75 min) induced apoptosis in isolated primary rat hepatocytes. The inhibitors (assigned with numbers) were: (1) Diphenyleneiodonium chloride (25 µM), (2) allopurinol (100 µM), (3) metyrapone (500 µM), (4) baicalein (5 µM), (5) AA861 (5 µM), (6) Indomethacine (10 µM), (7) nordihydroguaiaretic acid (20 µM), (8) arachidonyl trifluoromethylketone (5 µM), and (9) naringin (100 µM). After treatment, the cells were fixed evaluated for apoptosis and necrosis by microscopy. Treatment with nodularin gave rise to 86% apoptosis, compared to less than 1% in control.
Marinedrugs 13 06505 g002
The oxidative metabolism in mitochondria forms superoxide [68] that can be transported through voltage dependent anion channels to the cytosol [69] unless converted to freely diffusible H2O2 by mitochondrial superoxide dismutase. Mitochondria may not be the major primary target of CaM-KII mediated PP-inhibitor induced death as little impairment of nodularin-induced apoptosis was observed in hepatocytes pretreated (5 min) with inhibitors against mitochondrial complex I and III (rotenone and azide). Increased mitochondrial ROS has been reported in neurons undergoing OA-induced death [51]. Mitochondria can also be affected downstream of the ROS/CaM-KII-dependent activation of the apoptosis signaling kinase 1, which leads to activation of the c-Jun N-terminal Kinase and p38-MAPKinase apoptotic pathways [70,71,72,73].
The plasma membrane NADPH-oxidase generates superoxide, normally through stimulation by Rac [74], but also through stimulation by okadaic acid [75]. In our hands NADPH-oxidase inhibitor failed to prevent nodularin-induced hepatocyte apoptosis (Figure 2B).
The activity of oxygenases like xanthine oxidase (XO) and monoamine oxidases (MAO) can produce both peroxide and superoxide [76,77]. The aromatic amino acid hydroxylases form H2O2 when the oxidation of substrate is uncoupled [78,79], as when the enzyme binds the tetrahydrobiopterin cofactor without amino acid substrate [80]. It is of interest that CaM-KII phosphorylation of phenylalanine hydroxylase is enhanced in OA-exposed hepatocytes when the hydroxylase does not bind phenylalanine [81], suggesting that CaM-KII selectively activates the hydroxylase in a state prone to produce ROS rather than to hydroxylate phenylalanine.
Cytochrome P450 monooxygenases (CYP) can also generate ROS during conditions of poor coupling. The NADPH-CYP reductase and cytochrome b5 is a major source of ER-generated ROS [82]. Other known sources of ROS are nitric oxide synthase (NOS) [83] and enzymes of lipid metabolism such as lipoxygenases (LOXs) and cyclo-oxygenases (COXs) [84,85]. We have previously reported the involvement of CaM-KII and ROS in PPI-induced hepatocyte apoptosis [38,86]. To trace the source of the ROS we tested inhibitors of various ROS-producing enzymes, listed above, for ability to inhibit the nodularin-induced apoptosis of freshly isolated rat hepatocytes. The xanthine oxidase inhibitor allopurinol and the CYP inhibitor metyrapone interfered only slightly with the nodularin-induced apoptosis, while the NO synthase (NOS)/NADPH oxidase inhibitor diphenyleneiodonium, if anything, enhanced slightly the apoptosis (Figure 2B). We considered therefore if the nodularin could act by enhancing ROS production through the phospholipase A2/LOX/COX pathway. This pathway has been implicated in neuronal cell death [87], neurodegeneration and glutamate toxicity [88,89] and its superoxide production may be auto-amplified by ROS-induced phospholipase A2 (PLA2) activation [90].

9. The Potential Role of Perturbed Phospholipid Metabolism in PPI-Induced Hepatocyte Apoptosis

Inhibitors of cyclooxygenases COX1 and 2 (indomethacine) and against LOXs in general (nordihydroguaiaretic acid) were potent antagonists of nodularin-induced apoptosis. Interestingly, the 5-lipoxygenase (5-LOX) inhibitor AA861 protected against nodularin while the 12-lipoxygenase (12-LOX) inhibitor baicalein did not (Figure 2B). Although some of the inhibitors may have off-targets effects, like general anti-oxidant effects for nordihydroguaiaretic acid, the strong effect of all tested inhibitors of the PLA2/5-LOX/COX pathway is striking. These findings supplement the few previous reports connecting this pathway with PP inhibitors or CaM-KII [91,92].
It should be noted that okadaic acid enhances cellular CaM-KII catalyzed phosphorylation and activation of PLA2, which subsequently translocates from cytoplasm to perinuclear membranes [93], where it can co-localize with COX1 in the trans-Golgi network [94]. 5-LOX is presumably most active when complexed with the nuclear envelope and endoplasmic reticulum located 5-LOX activating protein (FLAP) and the leukotriene-C4 synthase [95] (see also [96] for a review). When phosphorylated by CaM-KII the 5-LOX can shift from a cytoplasmic to a nuclear location [97]. This may be significant because OA-induced cell death is counteracted by the perinuclearly located GTPase of the immune-associated proteins 5 (Gimap5) protein [98].

10. Gimap5 (Ian5, IROD) is an Inhibitor of OA-Induced Apoptosis

Gimap5, also known as Ian5 and IROD, is located in the perinuclear centrosomal/Golgi/ER compartment and its coiled-coil domain is essential for protection against both OA and acute ionizing radiation [98]. It was found by screening fibroblasts transduced with a protein expression library for survival at high OA concentration, and does not protect against microinjected cytochrome C, Fas-ligation, UV radiation, serum deprivation, or a number of chemotherapeutic drugs [98,99]. The major pathology in Gimap5 knock-out mice is decreased survival of T cells and natural killing cells as well as hepatocyte apoptosis and liver failure leading to death after about 15 weeks of life [100]. These findings imply that the perinuclear region where Gimap5 resides may be particularly vulnerable to the acute apoptotic death induced by PP inhibitors, also in hepatocytes, thus reinforcing the significance of the perinuclearly located ROS generating enzymes found required for PP inhibitor induced hepatocyte death (Figure 2B).

11. Concluding Remarks

In addition to their obvious health consequences, the marine protein phosphatase targeting toxins like okadaic acid give insight into cell death mechanisms. It is particularly intriguing that major actions of the protein phosphatase inhibiting toxins are mediated by CaM-KII in conjunction with increase of cellular ROS, possibly primarily in the perinuclear region of the target cell.

Acknowledgments

The authors greatly appreciate technical assistance from Nina Lied Larsen. This work was supported by the Norwegian Cancer Society, the Norwegian Research Council, and the Western Norway Regional Health Authorities (Helse Vest).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-LOX—5-lipoxygenase; 12-LOX—12-lipoxygenase; Adda—3-amino-9-methoxy-2,6,8-trimethyl-10-phenyl-4,6-decadienoic acid; AML—acute myeloid leukemia; Bcl—B-cell lymphoma; CaM-KII—Ca2+/calmodulin dependent protein kinase II; CalA—calyculin A; COX—cyclo-oxygenase; CYP—Cytochrome P450 complex; d-MeAsp—D-erythro-β-methylaspartic acid; Gimap5—GTPase of the immune associated protein family 5; MAO—monoamine oxidase; MAPK—mitogen activated protein kinase; MC—microcystin; Mdha—N-methyldehydroalanine; Mdhb—2-(methylamino)-2-dehydrobutyric acid; Nod—nodularin; NOX—NADPH oxidase; NOS—nitric oxide synthase; OA—okadaic acid; OATP—organic anion transporter polypeptide; PLA2—phospholipase A2; PP—protein phosphatase (of the phospho-Ser/-Thr phosphatase family); ROS—reactive oxygen species; XO—xanthine oxidase.

References

  1. Francis, G. Poisonous Australian lake. Nature 1878, 18, 11–12. [Google Scholar] [CrossRef]
  2. Heroes, E.; Lesage, B.; Gornemann, J.; Beullens, M.; van Meervelt, L.; Bollen, M. The PP1 binding code: A molecular-lego strategy that governs specificity. FEBS J. 2013, 280, 584–595. [Google Scholar] [CrossRef] [PubMed]
  3. Seshacharyulu, P.; Pandey, P.; Datta, K.; Batra, S.K. Phosphatase: PP2A structural importance, regulation and its aberrant expression in cancer. Cancer Lett. 2013, 335, 9–18. [Google Scholar] [CrossRef] [PubMed]
  4. Tachibana, K.; Scheuer, P.J.; Tsukitani, Y.; Kikuchi, H.; Vanengen, D.; Clardy, J.; Gopichand, Y.; Schmitz, F.J. Okadaic Acid, a Cyto-Toxic Polyether from 2 Marine Sponges of the Genus Halichondria. J. Am. Chem. Soc. 1981, 103, 2469–2471. [Google Scholar] [CrossRef]
  5. Bishop, C.T.; Anet, E.F.L.; Gorham, P.R. Isolation and Identification of the Fast-Death Factor in Microcystis aeruginosa NRC-1. Can. J. Biochem. Physiol. 1959, 37, 453–471. [Google Scholar] [PubMed]
  6. Botes, D.P.; Tuinman, A.A.; Wessels, P.L.; Viljoen, C.C.; Kruger, H.; Williams, D.H.; Santikarn, S.; Smith, R.J.; Hammond, S.J. The Structure of Cyanoginosin-LA, a Cyclic Heptapeptide Toxin from the Cyanobacterium Microcystis aeruginosa. J. Chem. Soc. Perkin Trans. 1 1984. [Google Scholar] [CrossRef]
  7. Carmichael, W.W.; Eschedor, J.T.; Patterson, G.M.L.; Moore, R.E. Toxicity and Partial Structure of a Hepatotoxic Peptide Produced by the Cyanobacterium Nodularia spumigena Mertens Emend L575 from New Zealand. Appl. Environ. Microbiol. 1988, 54, 2257–2263. [Google Scholar] [PubMed]
  8. Eriksson, J.E.; Meriluoto, J.A.O.; Kujari, H.P.; Osterlund, K.; Fagerlund, K.; Hallbom, L. Preliminary Characterization of a Toxin Isolated from the Cyanobacterium Nodularia spumigena. Toxicon 1988, 26, 161–166. [Google Scholar] [CrossRef]
  9. Rinehart, K.L.; Harada, K.; Namikoshi, M.; Chen, C.; Harvis, C.A.; Munro, M.H.G.; Blunt, J.W.; Mulligan, P.E.; Beasley, V.R.; Dahlem, A.M.; et al. Nodularin, Microcystin, and the Configuration of Adda. J. Am. Chem. Soc. 1988, 110, 8557–8558. [Google Scholar] [CrossRef]
  10. Connor, J.H.; Kleeman, T.; Barik, S.; Honkanen, R.E.; Shenolikar, S. Importance of the beta12-beta13 loop in protein phosphatase-1 catalytic subunit for inhibition by toxins and mammalian protein inhibitors. J. Biol. Chem. 1999, 274, 22366–22372. [Google Scholar] [CrossRef] [PubMed]
  11. Dawson, J.F.; Holmes, C.F. Molecular mechanisms underlying inhibition of protein phosphatases by marine toxins. Front. Biosci. 1999, 4, 646–658. [Google Scholar] [CrossRef]
  12. Gauss, C.M.; Sheppeck, J.E.; Nairn, A.C.; Chamberlin, R. A molecular modeling analysis of the binding interactions between the okadaic acid class of natural product inhibitors and the Ser-Thr phosphatases, PP1 and PP2A. Bioorg. Med. Chem. 1997, 5, 1751–1773. [Google Scholar] [CrossRef]
  13. Goldberg, J.; Huang, H.-b.; Kwon, Y.-g.; Greengard, P.; Nairn, A.C.; Kuriyan, J. Three-dimensional structure of the catalytic subunit of protein serine/threonine phosphatase-1. Nature 1995, 376, 745–753. [Google Scholar] [CrossRef] [PubMed]
  14. Maynes, J.T.; Bateman, K.S.; Cherney, M.M.; Das, A.K.; Luu, H.A.; Holmes, C.F.; James, M.N. Crystal structure of the tumor-promoter okadaic acid bound to protein phosphatase-1. J. Biol. Chem. 2001, 276, 44078–44082. [Google Scholar] [CrossRef] [PubMed]
  15. MacKintosh, R.W.; Dalby, K.N.; Campbell, D.G.; Cohen, P.T.W.; Cohen, P.; MacKintosh, C. The cyanobacterial toxin microcystin binds covalently to cysteine-273 on protein phosphatase 1. FEBS Lett. 1995, 371, 236–240. [Google Scholar] [CrossRef]
  16. Honkanen, R.E.; Dukelow, M.; Zwiller, J.; Moore, R.E.; Khatra, B.S.; Boynton, A.L. Cyanobacterial nodularin is a potent inhibitor of type 1 and type 2A protein phosphatases. Mol. Pharmacol. 1991, 40, 577–583. [Google Scholar] [PubMed]
  17. Swingle, M.; Honkanen, R.E. Small-molecule inhibitors of Ser/Thr protein phosphatases. In Methods in Molecular Biology; Moorhead, G., Ed.; Springer: New York, NY, USA, 2007; Volume 365, pp. 23–38. [Google Scholar]
  18. Honkanen, R.E.; Codispoti, B.A.; Tse, K.; Boynton, A.L. Characterization of natural toxins with inhibitory activity against serine/threonine protein phosphatases. Toxicon 1994, 32, 339–350. [Google Scholar] [CrossRef]
  19. MacKintosh, C.; Beattie, K.A.; Klumpp, S.; Cohen, P.; Codd, G.A. Cyanobacterial microcystin-LR is a potent and specific inhibitor of protein phosphatases 1 and 2A from both mammals and higher plants. FEBS Lett. 1990, 264, 187–192. [Google Scholar] [CrossRef]
  20. Toivola, D.M.; Eriksson, J.E.; Brautigan, D.L. Identification of protein phosphatase 2A as the primary target for microcystin-LR in rat liver homogenates. FEBS Lett. 1994, 344, 175–180. [Google Scholar] [CrossRef]
  21. Brewis, N.D.; Street, A.J.; Prescott, A.R.; Cohen, P.T. PPX, a novel protein serine/threonine phosphatase localized to centrosomes. EMBO J. 1993, 12, 987–996. [Google Scholar] [PubMed]
  22. Huang, X.; Honkanen, R.E. Molecular cloning, expression, and characterization of a novel human serine/threonine protein phosphatase, PP7, that is homologous to Drosophila retinal degeneration C gene product (rdgC). J. Biol. Chem. 1998, 273, 1462–1468. [Google Scholar] [CrossRef] [PubMed]
  23. Serres, M. H.; Fladmark, K. E.; Døskeland, S. O. An ultrasensitive competitive binding assay for the detection of toxins affecting protein phosphatases. Toxicon 2000, 38, 347–360. [Google Scholar] [CrossRef]
  24. Nam, K.Y.; Hiro, M.; Kimura, S.; Fujiki, H.; Imanishi, Y. Permeability of a Non-Tpa-Type Tumor Promoter, Okadaic Acid, through Lipid Bilayer-Membrane. Carcinogenesis 1990, 11, 1171–1174. [Google Scholar] [CrossRef] [PubMed]
  25. Becsi, B.; Kiss, A.; Erdodi, F. Interaction of protein phosphatase inhibitors with membrane lipids assessed by surface plasmon resonance based binding technique. Chem. Phys. Lipids 2014, 183, 68–76. [Google Scholar] [CrossRef] [PubMed]
  26. Oftedal, L.; Myhren, L.; Jokela, J.; Gausdal, G.; Sivonen, K.; Døskeland, S.O.; Herfindal, L. The lipopeptide toxins anabaenolysin A and B target biological membranes in a cholesterol-dependent manner. Biochim. Biophys. Acta 2012, 1818, 3000–3009. [Google Scholar] [CrossRef] [PubMed]
  27. Fischer, W.J.; Altheimer, S.; Cattori, V.; Meier, P.J.; Dietrich, D.R.; Hagenbuch, B. Organic anion transporting polypeptides expressed in liver and brain mediate uptake of microcystin. Toxicol. Appl. Pharmacol. 2005, 203, 257–263. [Google Scholar] [CrossRef] [PubMed]
  28. Lu, H.; Choudhuri, S.; Ogura, K.; Csanaky, I.L.; Lei, X.H.; Cheng, X.G.; Song, P.Z.; Klaassen, C.D. Characterization of organic anion transporting polypeptide 1b2-null mice: Essential role in hepatic uptake/toxicity of phalloidin and microcystin-LR. Toxicol. Sci. 2008, 103, 35–45. [Google Scholar] [CrossRef] [PubMed]
  29. Herfindal, L.; Kasprzykowski, F.; Schwede, F.; Lankiewicz, L.; Fladmark, K.E.; Lukomska, J.; Wahlsten, M.; Sivonen, K.; Grzonka, Z.; Jastorff, B.; et al. Acyloxymethyl esterification of nodularin-R and microcystin-LA produces inactive protoxins that become reactivated and produce apoptosis inside intact cells. J. Med. Chem. 2009, 52, 5758–5762. [Google Scholar] [CrossRef] [PubMed]
  30. Bøe, R.; Gjertsen, B.T.; Vintermyr, O.K.; Houge, G.; Lanotte, M.; Døskeland, S.O. The protein phosphatase inhibitor okadaic acid induces morphological changes typical of apoptosis in mammalian cells. Exp. Cell Res. 1991, 195, 237–246. [Google Scholar] [CrossRef]
  31. Herfindal, L. Bioactive Compounds from Culturable Marine Microorganisms. In Proceedings of the 5th International Conference on Bioprospecting, Tromso, Norway, 23–25 February 2011; p. 32.
  32. Komatsu, M.; Furukawa, T.; Ikeda, R.; Takumi, S.; Nong, Q.; Aoyama, K.; Akiyama, S.; Keppler, D.; Takeuchi, T. Involvement of mitogen-activated protein kinase signaling pathways in microcystin-LR-induced apoptosis after its selective uptake mediated by OATP1B1 and OATP1B3. Toxicol. Sci. 2007, 97, 407–416. [Google Scholar] [CrossRef] [PubMed]
  33. Monks, N.R.; Liu, S.; Xu, Y.; Yu, H.; Bendelow, A.S.; Moscow, J.A. Potent cytotoxicity of the phosphatase inhibitor microcystin LR and microcystin analogues in OATP1B1- and OATP1B3-expressing HeLa cells. Mol. Cancer Ther. 2007, 6, 587–598. [Google Scholar] [CrossRef] [PubMed]
  34. Fladmark, K.E.; Serres, M.H.; Larsen, N.L.; Yasumoto, T.; Aune, T.; Døskeland, S.O. Sensitive detection of apoptogenic toxins in suspension cultures of rat and salmon hepatocytes. Toxicon 1998, 36, 1101–1114. [Google Scholar] [CrossRef]
  35. Liu, L.; Jokela, J.; Herfindal, L.; Wahlsten, M.; Sinkkonen, J.; Permi, P.; Fewer, D.P.; Døskeland, S.O.; Sivonen, K. 4-Methylproline guided natural product discovery: Co-occurrence of 4-hydroxy- and 4-methylprolines in nostoweipeptins and nostopeptolides. ACS Chem. Biol. 2014, 9, 2646–2655. [Google Scholar] [CrossRef] [PubMed]
  36. Jokela, J.; Herfindal, L.; Wahlsten, M.; Permi, P.; Selheim, F.; Vasconcelos, V.; Døskeland, S.O.; Sivonen, K. A novel cyanobacterial nostocyclopeptide is a potent antitoxin against microcystin. ChemBioChem 2010, 11, 1594–1599. [Google Scholar] [CrossRef] [PubMed]
  37. Fladmark, K.E.; Brustugun, O.T.; Hovland, R.; Boe, R.; Gjertsen, B.T.; Zhivotovsky, B.; Døskeland, S.O. Ultrarapid caspase-3 dependent apoptosis induction by serine/threonine phosphatase inhibitors. Cell Death Differ. 1999, 6, 1099–1108. [Google Scholar] [CrossRef] [PubMed]
  38. Krakstad, C.; Herfindal, L.; Gjertsen, B.T.; Bøe, R.; Vintermyr, O.K.; Fladmark, K.E.; Døskeland, S.O. CaM-kinaseII dependent commitment to microcystin-induced apoptosis is coupled to cell budding, but not to shrinkage or chromatin hypercondensation. Cell Death Differ. 2006, 13, 1191–1202. [Google Scholar] [CrossRef] [PubMed]
  39. Houge, G.; Robaye, B.; Eikhom, T.S.; Golstein, J.; Mellgren, G.; Gjertsen, B.T.; Lanotte, M.; Døskeland, S.O. Fine mapping of 28S rRNA sites specifically cleaved in cells undergoing apoptosis. Mol. Cell. Biol. 1995, 15, 2051–2062. [Google Scholar] [CrossRef] [PubMed]
  40. Samali, A.; Gilje, B.; Doskeland, S.O.; Cotter, T.G.; Houge, G. The ability to cleave 28S ribosomal RNA during apoptosis is a cell-type dependent trait unrelated to DNA fragmentation. Cell Death Differ. 1997, 4, 289–293. [Google Scholar] [CrossRef] [PubMed]
  41. Vanden Berghe, T.; Linkermann, A.; Jouan-Lanhouet, S.; Walczak, H.; Vandenabeele, P. Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 2014, 15, 134–146. [Google Scholar]
  42. Gjertsen, B.T.; Cressey, L.I.; Ruchaud, S.; Houge, G.; Lanotte, M.; Doskeland, S.O. Multiple apoptotic death types triggered through activation of separate pathways by cAMP and inhibitors of protein phosphatases in one (IPC leukemia) cell line. J. Cell Sci. 1994, 107, 3363–3377. [Google Scholar] [PubMed]
  43. Pouria, S.; de Andrade, A.; Barbosa, J.; Cavalcanti, R.L.; Barreto, V.T.; Ward, C.J.; Preiser, W.; Poon, G.K.; Neild, G.H.; Codd, G.A. Fatal microcystin intoxication in haemodialysis unit in Caruaru, Brazil. Lancet 1998, 352, 21–26. [Google Scholar] [CrossRef]
  44. Jochimsen, E.M.; Carmichael, W.W.; An, J.S.; Cardo, D.M.; Cookson, S.T.; Holmes, C.E.M.; Antunes, M.B.D.; de Melo, D.A.; Lyra, T.M.; Barreto, V.S.T.; et al. Liver failure and death after exposure to microcystins at a hemodialysis center in Brazil. New Engl. J. Med. 1998, 338, 873–878. [Google Scholar] [CrossRef] [PubMed]
  45. Eriksson, J.E.; Paatero, G.I.; Meriluoto, J.A.; Codd, G.A.; Kass, G.E.; Nicotera, P.; Orrenius, S. Rapid microfilament reorganization induced in isolated rat hepatocytes by microcystin-LR, a cyclic peptide toxin. Exp. Cell Res. 1989, 185, 86–100. [Google Scholar] [CrossRef]
  46. Eriksson, J.E.; Toivola, D.; Meriluoto, J.A.; Karaki, H.; Han, Y.G.; Hartshorne, D. Hepatocyte deformation induced by cyanobacterial toxins reflects inhibition of protein phosphatases. Biochem. Biophys. Res. Commun. 1990, 173, 1347–1353. [Google Scholar] [CrossRef]
  47. Runnegar, M.T.; Falconer, I.R.; Silver, J. Deformation of isolated rat hepatocytes by a peptide hepatotoxin from the blue-green alga microcystis aeruginosa. Naunyn Schmiedebergs Arch. Pharmacol. 1981, 317, 268–272. [Google Scholar] [CrossRef] [PubMed]
  48. Toivola, D.M.; Goldman, R.D.; Garrod, D.R.; Eriksson, J.E. Protein phosphatases maintain the organization and structural interactions of hepatic keratin intermediate filaments. J. Cell Sci. 1997, 110, 23–33. [Google Scholar] [PubMed]
  49. Hirano, K.; Chartier, L.; Taylor, R.G.; Allen, R.E.; Fusetani, N.; Karaki, H.; Hartshorne, D.J. Changes in the cytoskeleton of 3T3 fibroblasts induced by the phosphatase inhibitor, calyculin-A. J. Muscle Res. Cell Motil. 1992, 13, 341–353. [Google Scholar] [CrossRef] [PubMed]
  50. Huseby, S.; Gausdal, G.; Keen, T.J.; Kjaerland, E.; Krakstad, C.; Myhren, L.; Bronstad, K.; Kunick, C.; Schwede, F.; Genieser, H.G.; et al. Cyclic AMP induces IPC leukemia cell apoptosis via CRE-and CDK-dependent Bim transcription. Cell Death Dis. 2011, 2, e237. [Google Scholar] [CrossRef] [PubMed]
  51. Cho, M.H.; Kim, D.H.; Choi, J.E.; Chang, E.J.; Yoon, S.Y. Increased phosphorylation of dynamin-related protein 1 and mitochondrial fission in okadaic acid-treated neurons. Brain Res. 2012, 1454, 100–110. [Google Scholar] [CrossRef] [PubMed]
  52. Fujiki, H.; Suganuma, M.; Suguri, H.; Yoshizawa, S.; Takagi, K.; Uda, N.; Wakamatsu, K.; Yamada, K.; Murata, M.; Yasumoto, T.; et al. Diarrhetic Shellfish Toxin, Dinophysistoxin-1, Is a Potent Tumor Promoter on Mouse Skin. Jpn. J. Cancer Res. 1988, 79, 1089–1093. [Google Scholar] [CrossRef] [PubMed]
  53. Nishiwaki-Matsushima, R.; Ohta, T.; Nishiwaki, S.; Suganuma, M.; Kohyama, K.; Ishikawa, T.; Carmichael, W.W.; Fujiki, H. Liver tumor promotion by the cyanobacterial cyclic peptide toxin microcystin-LR. J. Cancer Res. Clin. Oncol. 1992, 118, 420–424. [Google Scholar] [CrossRef] [PubMed]
  54. Pallas, D.C.; Shahrik, L.K.; Martin, B.L.; Jaspers, S.; Miller, T.B.; Brautigan, D.L.; Roberts, T.M. Polyoma Small and Middle T-Antigens and Sv40 Small T-Antigen Form Stable Complexes with Protein Phosphatase-2A. Cell 1990, 60, 167–176. [Google Scholar] [CrossRef]
  55. Yang, S.I.; Lickteig, R.L.; Estes, R.; Rundell, K.; Walter, G.; Mumby, M.C. Control of Protein Phosphatase-2A by Simian Virus-40 Small-T Antigen. Mol. Cell. Biol. 1991, 11, 1988–1995. [Google Scholar] [CrossRef] [PubMed]
  56. Harmala-Brasken, A.S.; Mikhailov, A.; Soderstrom, T.S.; Meinander, A.; Holmstrom, T.H.; Damuni, Z.; Eriksson, J.E. Type-2A protein phosphatase activity is required to maintain death receptor responsiveness. Oncogene 2003, 22, 7677–7686. [Google Scholar] [CrossRef] [PubMed]
  57. Mellgren, G.; Vintermyr, O.K.; Boe, R.; Døskeland, S.O. Hepatocyte DNA replication is abolished by inhibitors selecting protein phosphatase 2A rather than phosphatase 1. Exp. Cell Res. 1993, 205, 293–301. [Google Scholar] [CrossRef] [PubMed]
  58. Dean, N.M.; Mordan, L.J.; Tse, K.; Mooberry, S.L.; Boynton, A.L. Okadaic Acid Inhibits PDGF-Induced Proliferation and Decreases PDGF Receptor Number in C3H/10T1/2 Mouse Fibroblasts. Carcinogenesis 1991, 12, 665–670. [Google Scholar] [CrossRef] [PubMed]
  59. Gehringer, M.M. Microcystin-LR and okadaic acid-induced cellular effects: A dualistic response. FEBS Lett. 2004, 557, 1–8. [Google Scholar] [CrossRef]
  60. Nakagama, H.; Kaneko, S.; Shima, H.; Inamori, H.; Fukuda, H.; Kominami, R.; Sugimura, T.; Nagao, M. Induction of minisatellite mutation in NIH 3T3 cells by treatment with the tumor promoter okadaic acid. Proc. Natl. Acad. Sci. USA 1997, 94, 10813–10816. [Google Scholar] [CrossRef] [PubMed]
  61. Carvalho, P.S.; Catian, R.; Moukha, S.; Matias, W.G.; Creppy, E.E. Comparative study of Domoic Acid and Okadaic Acid induced-chromosomal abnormalities in the Caco-2 cell line. Int. J. Environ. Res. Public Health 2006, 3, 4–10. [Google Scholar] [CrossRef] [PubMed]
  62. Fladmark, K.E.; Brustugun, O.T.; Mellgren, G.; Krakstad, C.; Boe, R.; Vintermyr, O.K.; Schulman, H.; Døskeland, S.O. Ca2+/calmodulin-dependent protein kinase II is required for microcystin-induced apoptosis. J. Biol. Chem. 2002, 277, 2804–2811. [Google Scholar] [CrossRef] [PubMed]
  63. Coultrap, S.J.; Bayer, K.U. CaMKII regulation in information processing and storage. Trends Neurosci. 2012, 35, 607–618. [Google Scholar] [CrossRef] [PubMed]
  64. Erickson, J.R.; Joiner, M.L.A.; Guan, X.; Kutschke, W.; Yang, J.Y.; Oddis, C.V.; Bartlett, R.K.; Lowe, J.S.; O’Donnell, S.E.; Aykin-Burns, N.; et al. A dynamic pathway for calcium-independent activation of CaMKII by methionine oxidation. Cell 2008, 133, 462–474. [Google Scholar] [CrossRef] [PubMed]
  65. Denu, J.M.; Tanner, K.G. Specific and reversible inactivation of protein tyrosine phosphatases by hydrogen peroxide: Evidence for a sulfenic acid intermediate and implications for redox regulation. Biochemistry 1998, 37, 5633–5642. [Google Scholar] [CrossRef]
  66. Low, I.C.; Loh, T.; Huang, Y.; Virshup, D.M.; Pervaiz, S. Ser70 phosphorylation of Bcl-2 by selective tyrosine nitration of PP2A-B56delta stabilizes its antiapoptotic activity. Blood 2014, 124, 2223–2234. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, L.; Liu, L.; Yin, J.; Luo, Y.; Huang, S. Hydrogen peroxide-induced neuronal apoptosis is associated with inhibition of protein phosphatase 2A and 5, leading to activation of MAPK pathway. Int. J. Biochem. Cell Biol. 2009, 41, 1284–1295. [Google Scholar] [CrossRef] [PubMed]
  68. Drose, S.; Brandt, U. Molecular mechanisms of superoxide production by the mitochondrial respiratory chain. Adv. Exp. Med. Biol. 2012, 748, 145–169. [Google Scholar] [PubMed]
  69. Lustgarten, M.S.; Bhattacharya, A.; Muller, F.L.; Jang, Y.C.; Shimizu, T.; Shirasawa, T.; Richardson, A.; van Remmen, H. Complex I generated, mitochondrial matrix-directed superoxide is released from the mitochondria through voltage dependent anion channels. Biochem. Biophys. Res. Commun. 2012, 422, 515–521. [Google Scholar] [CrossRef] [PubMed]
  70. Fujino, G.; Noguchi, T.; Takeda, K.; Ichijo, H. Thioredoxin and protein kinases in redox signaling. Semin. Cancer Biol. 2006, 16, 427–435. [Google Scholar] [CrossRef] [PubMed]
  71. Saitoh, M.; Nishitoh, H.; Fujii, M.; Takeda, K.; Tobiume, K.; Sawada, Y.; Kawabata, M.; Miyazono, K.; Ichijo, H. Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. EMBO J. 1998, 17, 2596–2606. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, G.; Zhao, J.N.; Chang, Z.Y.; Guo, G.D. CaMKII Activates ASK1 to Induce Apoptosis of Spinal Astrocytes under Oxygen-Glucose Deprivation. Cell. Mol. Neurobiol. 2013, 33, 543–549. [Google Scholar] [CrossRef] [PubMed]
  73. Kashiwase, K.; Higuchi, Y.; Hirotani, S.; Yamaguchi, O.; Hikoso, S.; Takeda, T.; Watanabe, T.; Taniike, M.; Nakai, A.; Tsujimoto, I.; et al. CaMKII activates ASK1 and NF-kappa B to induce cardiomyocyte hypertrophy. Biochem. Biophys. Res. Commun. 2005, 327, 136–142. [Google Scholar] [CrossRef] [PubMed]
  74. Lorincz, A.M.; Szarvas, G.; Smith, S.M.; Ligeti, E. Role of Rac GTPase activating proteins in regulation of NADPH oxidase in human neutrophils. Free Radic. Biol. Med. 2014, 68, 65–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Steinbeck, M.J.; Hegg, G.G.; Karnovsky, M.J. Arachidonate Activation of the Neutrophil NADPH-Oxidase—Synergistic Effects of Protein Phosphatase Inhibitors Compared with Protein-Kinase Activators. J. Biol. Chem. 1991, 266, 16336–16342. [Google Scholar] [PubMed]
  76. Farquharson, C.A.; Butler, R.; Hill, A.; Belch, J.J.; Struthers, A.D. Allopurinol improves endothelial dysfunction in chronic heart failure. Circulation 2002, 106, 221–226. [Google Scholar] [CrossRef] [PubMed]
  77. Kaludercic, N.; Mialet-Perez, J.; Paolocci, N.; Parini, A.; Di Lisa, F. Monoamine oxidases as sources of oxidants in the heart. J. Mol. Cell. Cardiol. 2014, 73, 34–42. [Google Scholar] [CrossRef] [PubMed]
  78. Davis, M.D.; Kaufman, S. Products of the Tyrosine-Dependent Oxidation of Tetrahydrobiopterin by Rat-Liver Phenylalanine-Hydroxylase. Arch. Biochem. Biophys. 1993, 304, 9–16. [Google Scholar] [CrossRef] [PubMed]
  79. Haavik, J.; Almas, B.; Flatmark, T. Generation of reactive oxygen species by tyrosine hydroxylase: A possible contribution to the degeneration of dopaminergic neurons? J. Neurochem. 1997, 68, 328–332. [Google Scholar] [CrossRef] [PubMed]
  80. Adams, J.D.; Klaidman, L.K.; Ribeiro, P. Tyrosine hydroxylase: Mechanisms of oxygen radical formation. Redox. Rep. 1997, 3, 273–279. [Google Scholar] [PubMed]
  81. Doskeland, A.P.; Vintermyr, O.K.; Flatmark, T.; Cotton, R.G.H.; Doskeland, S.O. Phenylalanine Positively Modulates the cAMP-Dependent Phosphorylation and Negatively Modulates the Vasopressin-Induced and Okadaic-Acid-Induced Phosphorylation of Phenylalanine 4-Monooxygenase in Intact Rat Hepatocytes. Eur. J. Biochem. 1992, 206, 161–170. [Google Scholar] [CrossRef] [PubMed]
  82. Kim, H.R.; Lee, G.H.; Cho, E.Y.; Chae, S.W.; Ahn, T.; Chae, H.J. Bax inhibitor 1 regulates ER-stress-induced ROS accumulation through the regulation of cytochrome P450 2E1. J. Cell Sci. 2009, 122, 1126–1133. [Google Scholar] [CrossRef] [PubMed]
  83. Laursen, J.B.; Somers, M.; Kurz, S.; McCann, L.; Warnholtz, A.; Freeman, B.A.; Tarpey, M.; Fukai, T.; Harrison, D.G. Endothelial regulation of vasomotion in apoE-deficient mice: Implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation 2001, 103, 1282–1288. [Google Scholar] [CrossRef] [PubMed]
  84. Guidarelli, A.; Cantoni, O. Pivotal role of superoxides generated in the mitochondrial respiratory chain in peroxynitrite-dependent activation of phospholipase A2. Biochem. J. 2002, 366, 307–314. [Google Scholar] [CrossRef] [PubMed]
  85. Munoz, M.; Sanchez, A.; Pilar Martinez, M.; Benedito, S.; Lopez-Oliva, M.E.; Garcia-Sacristan, A.; Hernandez, M.; Prieto, D. COX-2 is involved in vascular oxidative stress and endothelial dysfunction of renal interlobar arteries from obese Zucker rats. Free Radic. Biol. Med. 2015, 84, 77–90. [Google Scholar] [CrossRef] [PubMed]
  86. Fladmark, K.E.; Brustugun, O.T.; Mellgren, G.; Krakstad, C.; Boe, R.; Vintermyr, O.K.; Schulman, H.; Doskeland, S.O. Ca2+/calmodulin-dependent protein kinase II is required for microcystin-induced apoptosis. J. Biol. Chem. 2002, 277, 2804–2811. [Google Scholar] [CrossRef] [PubMed]
  87. Im, J.Y.; Kim, D.; Paik, S.G.; Han, P.L. Cyclooxygenase-2-dependent neuronal death proceeds via superoxide anion generation. Free Radic. Biol. Med. 2006, 41, 960–972. [Google Scholar] [CrossRef] [PubMed]
  88. Kang, K.H.; Liou, H.H.; Hour, M.J.; Liou, H.C.; Fu, W.M. Protection of dopaminergic neurons by 5-lipoxygenase inhibitor. Neuropharmacology 2013, 73, 380–387. [Google Scholar] [CrossRef] [PubMed]
  89. Ha, J.S.; Dho, S.H.; Youm, T.H.; Kwon, K.S.; Park, S.S. Astrocytic phospholipase A2 contributes to neuronal glutamate toxicity. Brain Res. 2014, 1590, 97–106. [Google Scholar] [CrossRef] [PubMed]
  90. Zuo, L.; Christofi, F.L.; Wright, V.P.; Bao, S.; Clanton, T.L. Lipoxygenase-dependent superoxide release in skeletal muscle. J. Appl. Physiol. (1985) 2004, 97, 661–668. [Google Scholar] [CrossRef] [PubMed]
  91. Kanno, T.; Tsuchiya, A.; Shimizu, T.; Tanaka, A.; Nishizaki, T. Indomethacin Serves as a Potential Inhibitor of Protein Phosphatases. Cell Physiol. Biochem. 2012, 30, 1014–1022. [Google Scholar] [CrossRef] [PubMed]
  92. Ozveren, E.; Korkmaz, B.; Buharalioglu, C.K.; Tunctan, B. Involvement of calcium/calmodulin-dependent protein kinase II to endotoxin-induced vascular hyporeactivity in rat superior mesenteric artery. Pharmacol. Res. 2006, 54, 208–218. [Google Scholar] [CrossRef] [PubMed]
  93. Fatima, S.; Yaghini, F.A.; Ahmed, A.; Khandekar, Z.; Malik, K.U. CaM kinase II alpha mediates norepinephrine-induced translocation of cytosolic phospholipase A(2) to the nuclear envelope. J. Cell Sci. 2003, 116, 353–365. [Google Scholar] [CrossRef] [PubMed]
  94. Grewal, S.; Ponnambalam, S.; Walker, J.H. Association of cPLA(2)-alpha and COX-1 with the Golgi apparatus of A549 human lung epithelial cells. J. Cell Sci. 2003, 116, 2303–2310. [Google Scholar] [CrossRef] [PubMed]
  95. Strid, T.; Svartz, J.; Franck, N.; Hallin, E.; Ingelsson, B.; Soderstrom, M.; Hammarstrom, S. Distinct parts of leukotriene C-4 synthase interact with 5-lipoxygenase and 5-lipoxygenase activating protein. Biochem. Biophys. Res. Commun. 2009, 381, 518–522. [Google Scholar] [CrossRef] [PubMed]
  96. Radmark, O.; Werz, O.; Steinhilber, D.; Samuelsson, B. 5-Lipoxygenase: Regulation of expression and enzyme activity. Trends Biochem. Sci. 2007, 32, 332–341. [Google Scholar] [CrossRef] [PubMed]
  97. Fredman, G.; Ozcan, L.; Spolitu, S.; Hellmann, J.; Spite, M.; Backs, J.; Tabas, I. Resolvin D1 limits 5-lipoxygenase nuclear localization and leukotriene B-4 synthesis by inhibiting a calcium-activated kinase pathway. Proc. Natl. Acad. Sci. USA 2014, 111, 14530–14535. [Google Scholar] [CrossRef] [PubMed]
  98. Sandal, T.; Aumo, L.; Hedin, L.; Gjertsen, B.T.; Døskeland, S.O. Irod/Ian5: An inhibitor of gamma-radiation- and okadaic acid-induced apoptosis. Mol. Biol. Cell 2003, 14, 3292–3304. [Google Scholar] [CrossRef] [PubMed]
  99. Sandal, T.; Ahlgren, R.; Lillehaug, J.; Doskeland, S.O. Establishment of okadaic acid resistant cell clones using a cDNA expression library. Cell Death Differ. 2001, 8, 754–766. [Google Scholar] [CrossRef] [PubMed]
  100. Schulteis, R.D.; Chu, H.Y.; Dai, X.Z.; Chen, Y.H.; Edwards, B.; Haribhai, D.; Williams, C.B.; Malarkannan, S.; Hessner, M.J.; Glisic-Milosavljevic, S.; et al. Impaired survival of peripheral T cells, disrupted NK/NKT cell development, and liver failure in mice lacking Gimap5. Blood 2008, 112, 4905–4914. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Kleppe, R.; Herfindal, L.; Døskeland, S.O. Cell Death Inducing Microbial Protein Phosphatase Inhibitors—Mechanisms of Action. Mar. Drugs 2015, 13, 6505-6520. https://doi.org/10.3390/md13106505

AMA Style

Kleppe R, Herfindal L, Døskeland SO. Cell Death Inducing Microbial Protein Phosphatase Inhibitors—Mechanisms of Action. Marine Drugs. 2015; 13(10):6505-6520. https://doi.org/10.3390/md13106505

Chicago/Turabian Style

Kleppe, Rune, Lars Herfindal, and Stein Ove Døskeland. 2015. "Cell Death Inducing Microbial Protein Phosphatase Inhibitors—Mechanisms of Action" Marine Drugs 13, no. 10: 6505-6520. https://doi.org/10.3390/md13106505

Article Metrics

Back to TopTop