Effective PI3K modulators for improved therapy against malignant tumors and for neuroprotection of brain damage after tumor therapy (Review)

  • Authors:
    • Satoru Matsuda
    • Mayuko Ichimura
    • Mako Ogino
    • Noriko Nakano
    • Akari Minami
    • Toshiyuki Murai
    • Yasuko Kitagishi
  • View Affiliations

  • Published online on: September 27, 2016     https://doi.org/10.3892/ijo.2016.3710
  • Pages: 1785-1790
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Due to the key role in various cellular processes including cell proliferation and cell survival on many cell types, dysregulation of the PI3K/AKT pathway represents a crucial step of the pathogenesis in many diseases. Furthermore, the tumor suppressor PTEN negatively regulates the PI3K/AKT pathway through its lipid phosphatase activity, which is recognized as one of the most frequently deleted and/or mutated genes in human cancer. Given the pervasive involvement of this pathway, the development of the molecules that modulate this PI3K/AKT signaling has been initiated in studies which focus on the extensive effective drug discovery. Consequently, the PI3K/AKT pathway appears to be an attractive pharmacological target both for cancer therapy and for neurological protection necessary after the therapy. A better understanding of the molecular relations could reveal new targets for treatment development. We review recent studies on the features of PI3K/AKT and PTEN, and their pleiotropic functions relevant to the signaling pathways involved in cancer progress and in neuronal damage by the therapy.

1. Introduction

Phosphatidylinositol 3-kinase (PI3K)/AKT pathway is been known as an important signaling pathway in a variety of human cancers (13), which also plays critical roles in normal cell survival and proliferation under physiological condition (4,5). For example, prostate cancer development is often associated with silencing of the tumor suppressor phosphatase and tensin homolog (PTEN), a negative regulator of the PI3K/AKT signaling pathway (6,7). The PTEN is a multi-functional enzyme inhibiting the PI3K/AKT signaling in the cytosol and stabilizing genomes in the nucleus. Various extracellular signals including growth factors and/or cell nutrients could act as modulators for the PI3K/AKT/PTEN signaling axis. Once activated PI3K recruits cellular protein kinases such as the serine/threonine kinase AKT that consecutively transduces a signal to several downstream molecules. In general, relationship of the intracellular signaling networks coordinates cellular function containing cell cycle progression, proliferation and cell protection. Mutation in some of the effectors may result in consequent activation and/or inhibition of the signaling. This dysregulation of the signal occurs commonly in many malignancies, thereby rendering the cancer cells proliferative and with survival advantage. Accordingly, both nuclear and cytosolic PTEN/PI3K/AKT activity should be reflected for correlation with clinicopathological parameters of cancer (8). In addition, the PI3K/AKT signaling has been shown to induce the gene expression of multidrug resistance protein that is considerably associated with cancer therapy-opposition (9). Development of this multidrug resistance seems the major obstacle for chemotherapy of cancers. Therefore, it seems a remarkable target for molecular therapy in various diseases including cancer and related diseases. To date, many pharmacological inhibitors acting on the PI3K/AKT pathway have been established (10).

Overall, cancer treatment could have secondary negative effects such as neurocognitive deficits, serious sequelae that follow the therapy used to treat patients especially with brain neoplasms (11,12). The pathogenesis of the neurocognitive deficits involves apoptosis of neurons and other neuronal cells in a region in brain for learning and memory. After and during the intensive cancer therapy, brain damage has emerged as an important clinical problem (13,14). In regards to this, some reports show that nuclear trafficking of PTEN following brain injury leads to neuronal survival (15), indicating hope for new treatment options. The present review focuses on the recent advance in PI3K/AKT/PTEN-mediated neuroprotection and neurogenesis after several cancer therapies, highlighting its potential molecular and cellular mechanisms on the therapeutic advantages.

2. PI3K/AKT/PTEN pathway is involved in cancer development

Increased proliferation and cell motility are common cellular consequences associated with the high levels of intra-cellular phosphatidylinositol 3,4,5-triphosphate (PIP3) that give the metastatic characteristics to cancer (16). A feedback mechanism may increase the PIP3 further activating AKT and metastasis. Therefore, several studies have advocated that the PI3K/AKT signaling pathway is concerned with the progression and the prognosis of various types of cancer (17,18). Accordingly, suppression of PI3K and/or AKT inhibits proliferation and induces apoptosis in cancer cells (Fig. 1). On the contrary for example, dysregulation of the PI3K/AKT pathway seems to contribute to the malignant tumor activity with RET proto-oncogene mutations (19), which are involved in the pathogenesis of some forms of a neuroendocrine medullary thyroid cancer. In addition, it has also been shown that PI3K/AKT/mTOR signaling may play an important role in the biology of papillary tumors (20). Positive mTOR expression and PTEN loss may have a synergic effect on tumorigenesis and cancer cell proliferation (21). The evidence may lead to possible procedures of PI3K/AKT inhibitors in the therapy for patients with certain cancers (Fig. 1). In fact, targeting PI3K/AKT signaling pathway through specific inhibitors may represent an attractive potential therapeutic approach for the patients (19), actually exhibiting potent antitumor efficacy, providing rationale for clinical investigation of this inhibition in cancers of PI3K-mediated cells (22). Classically, wortmannin and LY294002 are the most characterized PI3K/AKT pathway inhibitors which prevent ATP from binding to the PI3Ks (23,24), both of which are cell-permeable and low molecular weight compounds. Wortmannin irreversibly inhibits PI3Ks, while the inhibition with LY294002 is reversible (10). Generally, PI3K inhibitors have been discovered to affect cell growth, proliferation and survival of cancer cells, as predicted before. The PI3K/AKT and its downstream inhibitors, when combined with other pharmacological agents would appear to be a more promising therapeutic modality. Particularly, PI3K/AKT inhibitors are prominently effective in PTEN null cells (25).

3. PI3K/AKT/PTEN pathway is involved in neurodegenerative disorders

ROS play a pivotal role in the pathogenesis both of neuroinflammatory and of neuro-degenerative diseases (26). Inflammatory conditions generally lead to the chronic neurodegenerative disease such as Parkinson's disease, Huntington's disease and Alzheimer's disease (27). Studies have indicated that cancer therapy commonly induces ROS production, which accelerates blood-brain barrier disruption and the neuronal cell death (28). Cancer therapy may induce apoptosis both for cancer cells and for normal neuronal cells. In this meaning, reduction of ROS may have a high potential to diminish brain damage induced during cancer therapy. It has been demonstrated that upregulation of PTEN causes modulation of PI3K/AKT signaling pathway to reduce the ROS generation in cells (29). As described before, the PTEN is one of the most prominent tumor suppressor genes that is frequently deleted or mutated in a variety of human cancers (30). In addition, PTEN is highly expressed in neurons and it has been described to be related to critical neuroprotection (31,32). Recently, it has been shown that AKT activation may likely play a therapeutic role in neurodegenerative diseases (33,34). Schematic structures of the AKT and PTEN protein are shown in Fig. 2. Accordingly, the tumor suppressor PTEN, which antagonizes the PI3K/AKT pathway, has been recognized to play a crucial role in neural functions. Neuroprotection by inhibiting PTEN has been reported by activating the PI3K/AKT pathway in primary neurons (35,36). Deletion of PTEN gene has been shown to result in cognitive impairment (37). Ischemic stroke induces rapid PTEN degradation in both neurons and astrocytes which play both protective and detrimental action to neurons in a spatiotemporal- and cell-type-dependent manner (38). Simultaneous deletion of PTEN promotes significant nerve regeneration after the crush injury with enhanced axon regeneration (39). However, there is somewhat controversy whether accumulation of the tumor suppressor PTEN protein under stress conditions such as trauma and stroke causes neuronal cell death. A number of studies have reported enhanced apoptosis in neurons possessing increased amount of nuclear PTEN, with the interpretation that its nuclear phosphatase activity leads to the reduction of survival AKT signaling activity. One interpretation is that brain trauma may modify the nucleo-cytoplasmic distribution of PTEN, resulting in the increased nuclear PTEN, but only in surviving neurons near the traumatic lesion (15,40).

4. Potential therapeutic approach for neuro-cellular protection via the modulation of PI3K/AKT/PTEN pathway

Cognitive failures after brain tumor and its treatments are appropriately consistent, although there is only a small worldwide body of research describing them (41). Chemotherapy against cancer is often associated with cognitive deficits which may remain after the end of the treatment (42). In the central nervous system, PI3K/AKT signaling modifies synaptic plasticity underlying the memory-processes suggesting that PI3K/AKT signaling contributes to improvement in the cognitive development. Inhibition of PTEN is expected to promote neural cell survival, neuroprotection and neuroregeneration, which can also promote myelination of axons through the AKT activation (43). Actually, a PTEN inhibitor reduces tissue damage, neuronal cell death and promotes the functional recovery (43). In addition, it has been shown that PTEN-induced putative kinase 1 (PINK1) induces mitophagy promoting neuroprotection on Huntington's disease (44). Furthermore, anti-depressants acting on serotonin neurotransmission generally activate AKT then inhibit the downstream GSK3β in neuronal cells. Several psychoactive medicines have been shown to modulate the activity of the PI3K/AKT signaling (45) (Fig. 2). AKT has downstream substrates such as the GSK3β (46). MAO inhibitors and SSRI antidepressants which promote serotonin synaptic transmission may inhibit the GSK3β activity (47). Atypical antipsychotics may inhibit the regulation of the GSK3β signaling (48). Similar results have been reported after the treatment with haloperidol (49,50). Chlorpromazine is a prominent antipsychotic agent initially developed to control several psychotic disorders, which also inhibits cell proliferation and cell survival by suppressing PI3K/AKT signaling pathway (51). Whereas AKT pathway is regulated by different types of psychiatric drugs in this way, lithium activates PI3K and PI3K-dependent downstream AKT signaling (52). These activities eventually protect neuronal cells against neuronal toxicity. Therefore, treatment with inhibitors of PI3K may exacerbate neuronal cell death after brain damage in the area of injury. On the contrary, inhibition of PTEN expression diminishes the neurological damage after brain injury. In addition, it has been revealed that pharmacological inhibition of PTEN protects against brain injury in a dose-dependent manner, whose protective effect might be induced via the upregulation of PI3K/AKT signaling (53,54). Furthermore, PTEN has some crosstalk effects with serotonin receptor signaling (55). By blocking dopamine D2 receptors, classic antipsychotics can prevent the inhibition of AKT signaling (56,57). In addition, reductions in AKT activation in neurons may increase excitability through decreases in GABA neurotransmission (58). It seems important to exploit the potential benefits of these optimal treatments and/or combination with the PI3K/AKT modulators for the actual neuroprotection against neurological damage.

5. Dietary regulation of PI3K/AKT signaling via the modified PTEN expression

The PI3K/AKT and PTEN signaling plays various cellular key roles under normal and/or pathological conditions. Activation of both pathways can impact functional consequences in cancer therapy and the subsequent multiple CNS disorders. Of course, pharmacological interference of signaling molecules provides convenient method. For example, bisperoxovanadium compounds inhibit PTEN signaling, and have been used for elevation of neuroprotection in many CNS damage studies (43,59,60). In addition, small molecules have prevalent use by means of experimental tools as well as therapeutics (61). Now, it is challenging to describe appropriate strategies to achieve cost effective benefits from easy diets to control the PTEN signaling molecules. Dietary and/or therapeutic interventions could not only contribute to the prevention of diseases but reduce the rate of its development. Actually, several herbs had been established to have characteristics for antitumor activity (62). In addition, dietary regulation of PI3K/AKT signaling via the modulating PTEN expression might be effective both for cancer inhibition and for a neuroprotection. For example, dietary intake of isothiocyanate sulforaphane, a dietary isothiocyanate derived from broccoli, modulates PTEN gene expression (63). Dietary exposure to the soy isoflavone such as genistein also induces PTEN expression at physiologically relevant concentrations (64). Phytoestrogen exposure might result in an increase in PTEN expression and the subsequent decrease in cellular responses including AKT phosphorylation. Accordingly, consumption of moderate levels of soy, vegetables, some fruits, and red wine can be protective against cancers. A high-fat diet raises circulating fatty acids, which also increases PTEN expression in prostate epithelial cells (65). In addition, dietary intake of indole-3-carbinol upregulates PTEN in an animal model (66). Indole-3-carbinol is a promising cancer-preventive phytochemical found in some vegetables such as broccoli. PTEN expression at the level of mRNA and protein is elevated in experimental animals fed with whey protein (67) which has been shown to possess multiple health benefits (67). In addition, it has been reported that DHA and EPA raise the level of PTEN in breast cancer cells, providing a mechanism for the beneficial effects of fish oils on breast cancer cells (68,69). Fish oil rich in polyunsaturated fatty acids may induce the PTEN expression by activation of peroxisome proliferator-activated receptor (PPAR) (68,69), which also attenuate neuron cellular damage after a brain ischemia and appear to play an important role in the activation of anti-apoptotic signaling (70). Schematic structures of human PPAR and PTEN are shown in Fig. 3. PUFA ethanolamides DHA and EPA induce autophagy through PPAR activation in cancer cells (68). In contrast, high fat diet attenuates the neuroprotection because of decreased survival of AKT signaling (71). A lignan Honokiol isolated from the bark of Magnolia officinalis could attenuate PI3K/AKT signaling by upregulation of PTEN expression (72), which is a potential antitumor compound. Honokiol has been reported to improve the learning and memory impairments in experimental animals (73). A food ingredient curcumin, derived from the root of the plant Curcuma longa, repairs PTEN expression (74). Curcumin inhibits cell proliferation in human osteoclastoma cells (75). Furthermore, curcumin is a potential therapeutic mediator for neuro-cognition (76). In contrast, certain component of rosemary herb decreases the expression of PTEN in K562 human myeloid cells (77). Resveratrol, an ingredient in grapes, has been reported to exhibit anticancer activity, anti-inflammatory activity and cardiovascular protection property. Notably, resveratrol has been recently reported to have neuroprotective effect (78). Supplementation of these natural compounds may provide an innovative therapeutic approach to brain disorders after cancer therapy (31).

6. So what next?

Increasing evidence pointing to AKT pathway-modification in psychotic disorders offer a novel implication of the treatment mechanisms for rescuing brain damage after cancer therapy. Although PTEN has been discovered as a tumor suppressor, PTEN is also involved in several other diseases, and may be regulated at multiple levels including transcription, protein stability and phosphorylation. Potential synergy with other targeted inhibitors and/or with conventional chemotherapy may provide additional therapeutic options to optimize the therapeutic efficacy. In addition, abnormalities in the PI3K/AKT signaling pathway and in the cross-talk pathways provide a clear rationale for the development of signal transduction inhibitor-based approaches aimed for both cancer therapy and the subsequent neurological disorders. Understanding the important roles of the intracellular signaling and the intricate internet work communication is an important direction for making effective and specific therapies. However, much more work is needed to effectively produce and validate therapies that target pathways. Particularly, it will be now a challenge to seek out how to use medicinal herbs for the correction in critical processes required for maintaining neuronal homeostasis linking to a brain injury characterized by cancer therapy. Current evidence, as described here, suggests that work toward understanding these complex signaling cascades is not only promising, but may be critical for achieving the goal of improved neurological outcome after brain and spinal cord damage. This relationship has similarities with another relationship between methotrexate (MTX) therapy and a leucovorin (LV) rescue (79). We think the order of the usage of medicine and/or compound for the treatment is the most important. Of course, precise understanding of these regulations is crucial for therapeutic intervention and the effective design of novel therapeutics. A comprehensive description of the PI3K/AKT signaling network has to be evolved. Studies may be ongoing to identify the most optimal inhibitors for cancer subtypes.

Acknowledgements

The present study was supported by the JSPS KAKENHI grant number 26-12035, 24240098.

Abbreviations:

BDNF

brain-derived neurotrophic factor

DHA

docosahexaenoic acid

EPA

eicosapentaenoic acid

FABP4

fattyacid-binding protein 4

GABA

gamma-aminobutyric acid

GSK3

glycogen synthase kinase 3

HSF1

heat shock factor 1

5-HT

5-hydroxytryptamine, serotonin

mTOR

mammalian target of rapamycin

PIP3

phosphatidylinositol 3,4,5-triphosphate

PIP2

phosphatidylinositol 4,5-bisphosphate

PI3K

phosphoinositide-3 kinase

PTEN

phosphatase and tensin homologue deleted on chromosome 10

PPAR

peroxisome proliferator-activated receptor

PUFAs

polyunsaturated fatty acids

ROS

reactive oxygen species

SSRI

selective serotonin reuptake inhibitors

References

1 

Lee JJ, Loh K and Yap YS: PI3K/Akt/mTOR inhibitors in breast cancer. Cancer Biol Med. 12:342–354. 2015.

2 

Robbins HL and Hague A: The PI3K/Akt pathway in tumors of endocrine tissues. Front Endocrinol (Lausanne). 6:1882016.

3 

Faes S and Dormond O: PI3K and AKT: Unfaithful partners in cancer. Int J Mol Sci. 16:21138–21152. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Gesmundo I, Villanova T, Gargantini E, Arvat E, Ghigo E and Granata R: The mineralocorticoid agonist fludrocortisone promotes survival and proliferation of adult hippocampal progenitors. Front Endocrinol (Lausanne). 7:662016.

5 

Wahane SD, Hellbach N, Prentzell MT, Weise SC, Vezzali R, Kreutz C, Timmer J, Krieglstein K, Thedieck K and Vogel T: PI3K-p110-alpha-subtype signalling mediates survival, proliferation and neurogenesis of cortical progenitor cells via activation of mTORC2. J Neurochem. 130:255–267. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Simper NB, Jones CL, MacLennan GT, Montironi R, Williamson SR, Osunkoya AO, Wang M, Zhang S, Grignon DJ, Eble JN, et al: Basal cell carcinoma of the prostate is an aggressive tumor with frequent loss of PTEN expression and overexpression of EGFR. Hum Pathol. 46:805–812. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Fallahabadi ZR, Noori Daloii MR, Mahdian R, Behjati F, Shokrgozar MA, Abolhasani M, Asgari M and Shahrokh H: Frequency of PTEN alterations, TMPRSS2-ERG fusion and their association in prostate cancer. Gene. 575:755–760. 2016. View Article : Google Scholar

8 

Collaud S, Tischler V, Atanassoff A, Wiedl T, Komminoth P, Oehlschlegel C, Weder W and Soltermann A: Lung neuroendocrine tumors: Correlation of ubiquitinylation and sumoylation with nucleo-cytosolic partitioning of PTEN. BMC Cancer. 15:742015. View Article : Google Scholar : PubMed/NCBI

9 

Chen SY, Zheng XW, Cai JX, Zhang WP, You HS, Xing JF and Dong YL: Histone deacetylase inhibitor reverses multidrug resistance by attenuating the nucleophosmin level through PI3K/Akt pathway in breast cancer. Int J Oncol. 49:294–304. 2016.PubMed/NCBI

10 

Sinagra T, Tamburella A, Urso V, Siarkos I, Drago F, Bucolo C and Salomone S: Reversible inhibition of vasoconstriction by thiazolidinediones related to PI3K/Akt inhibition in vascular smooth muscle cells. Biochem Pharmacol. 85:551–559. 2013. View Article : Google Scholar

11 

Day J, Gillespie DC, Rooney AG, Bulbeck HJ, Zienius K, Boele F and Grant R: Neurocognitive deficits and neurocognitive rehabilitation in adult brain tumors. Curr Treat Options Neurol. 18:222016. View Article : Google Scholar : PubMed/NCBI

12 

Cheung YT and Krull KR: Neurocognitive outcomes in long-term survivors of childhood acute lymphoblastic leukemia treated on contemporary treatment protocols: A systematic review. Neurosci Biobehav Rev. 53:108–120. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Zheng Q, Yang L, Tan LM, Qin LX, Wang CY and Zhang HN: Stroke-like migraine attacks after radiation therapy syndrome. Chin Med J (Engl). 128:2097–2101. 2015. View Article : Google Scholar

14 

Ricard D, Taillia H and Renard JL: Brain damage from anticancer treatments in adults. Curr Opin Oncol. 21:559–565. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Goh CP, Putz U, Howitt J, Low LH, Gunnersen J, Bye N, Morganti-Kossmann C and Tan SS: Nuclear trafficking of Pten after brain injury leads to neuron survival not death. Exp Neurol. 252:37–46. 2014. View Article : Google Scholar

16 

Qin J, Xie Y, Wang B, Hoshino M, Wolff DW, Zhao J, Scofield MA, Dowd FJ, Lin MF and Tu Y: Upregulation of PIP3-dependent Rac exchanger 1 (P-Rex1) promotes prostate cancer metastasis. Oncogene. 28:1853–1863. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Chen F, Chen X, Yang D, Che X, Wang J, Li X, Zhang Z, Wang Q, Zheng W, Wang L, et al: Isoquercitrin inhibits bladder cancer progression in vivo and in vitro by regulating the PI3K/Akt and PKC signaling pathways. Oncol Rep. 36:165–172. 2016.PubMed/NCBI

18 

Zhang J, Yu XH, Yan YG, Wang C and Wang WJ: PI3K/Akt signaling in osteosarcoma. Clin Chim Acta. 444:182–192. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Manfredi GI, Dicitore A, Gaudenzi G, Caraglia M, Persani L and Vitale G: PI3K/Akt/mTOR signaling in medullary thyroid cancer: A promising molecular target for cancer therapy. Endocrine. 48:363–370. 2015. View Article : Google Scholar

20 

Goschzik T, Gessi M, Denkhaus D and Pietsch T: PTEN mutations and activation of the PI3K/Akt/mTOR signaling pathway in papillary tumors of the pineal region. J Neuropathol Exp Neurol. 73:747–751. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Han X, Ji Y, Zhao J, Xu X and Lou W: Expression of PTEN and mTOR in pancreatic neuroendocrine tumors. Tumour Biol. 34:2871–2879. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Lee M, Wiedemann T, Gross C, Leinhäuser I, Roncaroli F, Braren R and Pellegata NS: Targeting PI3K/mTOR signaling displays potent antitumor efficacy against nonfunctioning pituitary adenomas. Clin Cancer Res. 21:3204–3215. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Bony C, Roche S, Shuichi U, Sasaki T, Crackower MA, Penninger J, Mano H and Pucéat M: A specific role of phosphatidylinositol 3-kinase gamma. A regulation of autonomic Ca2+ oscillations in cardiac cells. J Cell Biol. 152:717–728. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Yanamandra M, Mitra S and Giri A: Development and application of PI3K assays for novel drug discovery. Expert Opin Drug Discov. 10:171–186. 2015. View Article : Google Scholar

25 

Lassen A, Atefi M, Robert L, Wong DJ, Cerniglia M, Comin-Anduix B and Ribas A: Effects of AKT inhibitor therapy in response and resistance to BRAF inhibition in melanoma. Mol Cancer. 13:832014. View Article : Google Scholar : PubMed/NCBI

26 

Schilling T and Eder C: Stimulus-dependent requirement of ion channels for microglial NADPH oxidase-mediated production of reactive oxygen species. J Neuroimmunol. 225:190–194. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Clark LF and Kodadek T: The immune system and neuroinflammation as potential sources of blood-based biomarkers for Alzheimer's disease, Parkinson's disease, and Huntington's disease. ACS Chem Neurosci. 7:520–527. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Lam M, Oleinick NL and Nieminen AL: Photodynamic therapy-induced apoptosis in epidermoid carcinoma cells. Reactive oxygen species and mitochondrial inner membrane permeabilization. J Biol Chem. 276:47379–47386. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Zhu Y, Hoell P, Ahlemeyer B, Sure U, Bertalanffy H and Krieglstein J: Implication of PTEN in production of reactive oxygen species and neuronal death in in vitro models of stroke and Parkinson's disease. Neurochem Int. 50:507–516. 2007. View Article : Google Scholar

30 

Lim HJ, Crowe P and Yang JL: Current clinical regulation of PI3K/PTEN/Akt/mTOR signalling in treatment of human cancer. J Cancer Res Clin Oncol. 141:671–689. 2015. View Article : Google Scholar

31 

Kitagishi Y and Matsuda S: Diets involved in PPAR and PI3K/AKT/PTEN pathway may contribute to neuroprotection in a traumatic brain injury. Alzheimers Res Ther. 5:422013. View Article : Google Scholar : PubMed/NCBI

32 

Liu NK and Xu XM: Neuroprotection and its molecular mechanism following spinal cord injury. Neural Regen Res. 7:2051–2062. 2012.PubMed/NCBI

33 

Cheng B, Martinez AA, Morado J, Scofield V, Roberts JL and Maffi SK: Retinoic acid protects against proteasome inhibition associated cell death in SH-SY5Y cells via the AKT pathway. Neurochem Int. 62:31–42. 2013. View Article : Google Scholar

34 

Ermak G, Hench KJ, Chang KT, Sachdev S and Davies KJ: Regulator of calcineurin (RCAN1-1L) is deficient in Huntington disease and protective against mutant huntingtin toxicity in vitro. J Biol Chem. 284:11845–11853. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Walker CL, Liu NK and Xu XM: PTEN/PI3K and MAPK signaling in protection and pathology following CNS injuries. Front Biol (Beijing). 8:421–433. 2013. View Article : Google Scholar

36 

Delgado-Esteban M, Martin-Zanca D, Andres-Martin L, Almeida A and Bolaños JP: Inhibition of PTEN by peroxynitrite activates the phosphoinositide-3-kinase/Akt neuroprotective signaling pathway. J Neurochem. 102:194–205. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Takeuchi K, Gertner MJ, Zhou J, Parada LF, Bennett MV and Zukin RS: Dysregulation of synaptic plasticity precedes appearance of morphological defects in a Pten conditional knockout mouse model of autism. Proc Natl Acad Sci USA. 110:4738–4743. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Li W, Huang R, Chen Z, Yan LJ, Simpkins JW and Yang SH: PTEN degradation after ischemic stroke: A double-edged sword. Neuroscience. 274:153–161. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Sun F, Park KK, Belin S, Wang D, Lu T, Chen G, Zhang K, Yeung C, Feng G, Yankner BA, et al: Sustained axon regeneration induced by co-deletion of PTEN and SOCS3. Nature. 480:372–375. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Li Y, Low LH, Putz U, Goh CP, Tan SS and Howitt J: Rab5 and Ndfip1 are involved in Pten ubiquitination and nuclear trafficking. Traffic. 15:749–761. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Mandelblatt JS, Hurria A, McDonald BC, Saykin AJ, Stern RA, VanMeter JW, McGuckin M, Traina T, Denduluri N, Turner S, et al; Thinking and Living With Cancer Study. Cognitive effects of cancer and its treatments at the intersection of aging: What do we know; what do we need to know? Semin Oncol. 40:709–725. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Evenden J: Cognitive impairments and cancer chemotherapy: Translational research at a crossroads. Life Sci. 93:589–595. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Walker CL and Xu XM: PTEN inhibitor bisperoxovanadium protects oligodendrocytes and myelin and prevents neuronal atrophy in adult rats following cervical hemicontusive spinal cord injury. Neurosci Lett. 573:64–68. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Khalil B, El Fissi N, Aouane A, Cabirol-Pol MJ, Rival T and Liévens JC: PINK1-induced mitophagy promotes neuroprotection in Huntington's disease. Cell Death Dis. 6:e16172015. View Article : Google Scholar : PubMed/NCBI

45 

Ozaita A, Puighermanal E and Maldonado R: Regulation of PI3K/Akt/GSK-3 pathway by cannabinoids in the brain. J Neurochem. 102:1105–1114. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Li S, Zhou Y, Wang R, Zhang H, Dong Y and Ip C: Selenium sensitizes MCF-7 breast cancer cells to doxorubicin-induced apoptosis through modulation of phospho-Akt and its downstream substrates. Mol Cancer Ther. 6:1031–1038. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Hui J, Zhang J, Kim H, Tong C, Ying Q, Li Z, Mao X, Shi G, Yan J, Zhang Z, et al: Fluoxetine regulates neurogenesis in vitro through modulation of GSK-3β/β-catenin signaling. Int J Neuropsychopharmacol. 18:pyu0992014. View Article : Google Scholar

48 

Park SW, Phuong VT, Lee CH, Lee JG, Seo MK, Cho HY, Fang ZH, Lee BJ and Kim YH: Effects of antipsychotic drugs on BDNF, GSK-3β, and β-catenin expression in rats subjected to immobilization stress. Neurosci Res. 71:335–340. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Emamian ES, Hall D, Birnbaum MJ, Karayiorgou M and Gogos JA: Convergent evidence for impaired AKT1-GSK3beta signaling in schizophrenia. Nat Genet. 36:131–137. 2004. View Article : Google Scholar : PubMed/NCBI

50 

Kozlovsky N, Amar S, Belmaker RH and Agam G: Psychotropic drugs affect Ser9-phosphorylated GSK-3 beta protein levels in rodent frontal cortex. Int J Neuropsychopharmacol. 9:337–342. 2006. View Article : Google Scholar

51 

Shin SY, Lee KS, Choi YK, Lim HJ, Lee HG, Lim Y and Lee YH: The antipsychotic agent chlorpromazine induces autophagic cell death by inhibiting the Akt/mTOR pathway in human U-87MG glioma cells. Carcinogenesis. 34:2080–2089. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Liu KJ, Lee YL, Yang YY, Shih NY, Ho CC, Wu YC, Huang TS, Huang MC, Liu HC, Shen WW, et al: Modulation of the development of human monocyte-derived dendritic cells by lithium chloride. J Cell Physiol. 226:424–433. 2011. View Article : Google Scholar

53 

Wu J, Li J, Hu H, Liu P, Fang Y and Wu D: Rho-kinase inhibitor, fasudil, prevents neuronal apoptosis via the Akt activation and PTEN inactivation in the ischemic penumbra of rat brain. Cell Mol Neurobiol. 32:1187–1197. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Liu B, Li L, Zhang Q, Chang N, Wang D, Shan Y, Li L, Wang H, Feng H, Zhang L, et al: Preservation of GABAA receptor function by PTEN inhibition protects against neuronal death in ischemic stroke. Stroke. 41:1018–1026. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Cai J, Yi Z, Lu W, Fang Y and Zhang C: Crosstalk between 5-HT2cR and PTEN signaling pathway in atypical antipsychotic-induced metabolic syndrome and cognitive dysfunction. Med Hypotheses. 80:486–489. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Mirones I, Angel Rodríguez-Milla M, Cubillo I, Mariñas-Pardo L, de la Cueva T, Zapata A, González C, Ramírez M and García-Castro J: Dopamine mobilizes mesenchymal progenitor cells through D2-class receptors and their PI3K/AKT pathway. Stem Cells. 32:2529–2538. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Stavarache MA, Musatov S, McGill M, Vernov M and Kaplitt MG: The tumor suppressor PTEN regulates motor responses to striatal dopamine in normal and Parkinsonian animals. Neurobiol Dis. 82:487–494. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Souza BR, Romano-Silva MA and Tropepe V: Dopamine D2 receptor activity modulates Akt signaling and alters GABAergic neuron development and motor behavior in zebrafish larvae. J Neurosci. 31:5512–5525. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Walker CL, Walker MJ, Liu NK, Risberg EC, Gao X, Chen J and Xu XM: Systemic bisperoxovanadium activates Akt/mTOR, reduces autophagy, and enhances recovery following cervical spinal cord injury. PLoS One. 7:e300122012. View Article : Google Scholar : PubMed/NCBI

60 

Mao D and Sun X: Reactivation of the PI3K/Akt signaling pathway by the bisperoxovanadium compound bpV(pic) attenuates photoreceptor apoptosis in experimental retinal detachment. Invest Ophthalmol Vis Sci. 56:5519–5532. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Spinelli L, Lindsay YE and Leslie NR: PTEN inhibitors: an evaluation of current compounds. Adv Biol Regul. 57:102–111. 2015. View Article : Google Scholar

62 

Tian HY, Li ZX, Li HY, Wang HJ, Zhu XW and Dou ZH: Effects of 14 single herbs on the induction of caspase-3 in tumor cells: A brief review. Chin J Integr Med. 19:636–640. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Traka MH, Spinks CA, Doleman JF, Melchini A, Ball RY, Mills RD and Mithen RF: The dietary isothiocyanate sulforaphane modulates gene expression and alternative gene splicing in a PTEN null preclinical murine model of prostate cancer. Mol Cancer. 9:1892010. View Article : Google Scholar : PubMed/NCBI

64 

de la Parra C, Castillo-Pichardo L, Cruz-Collazo A, Cubano L, Redis R, Calin GA and Dharmawardhane S: Soy isoflavone genistein-mediated downregulation of miR-155 contributes to the anticancer effects of genistein. Nutr Cancer. 68:154–164. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Kwon OJ, Zhang B, Zhang L and Xin L: High fat diet promotes prostatic basal-to-luminal differentiation and accelerates initiation of prostate epithelial hyperplasia originated from basal cells. Stem Cell Res (Amst). 16:682–691. 2016. View Article : Google Scholar

66 

Aronchik I, Kundu A, Quirit JG and Firestone GL: The antiproliferative response of indole-3-carbinol in human melanoma cells is triggered by an interaction with NEDD4-1 and disruption of wild-type PTEN degradation. Mol Cancer Res. 12:1621–1634. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Eason RR, Velarde MC, Chatman L Jr, Till SR, Geng Y, Ferguson M, Badger TM and Simmen RC: Dietary exposure to whey proteins alters rat mammary gland proliferation, apoptosis, and gene expression during postnatal development. J Nutr. 134:3370–3377. 2004.PubMed/NCBI

68 

Rovito D, Giordano C, Vizza D, Plastina P, Barone I, Casaburi I, Lanzino M, De Amicis F, Sisci D, Mauro L, et al: Omega-3 PUFA ethanolamides DHEA and EPEA induce autophagy through PPARγ activation in MCF-7 breast cancer cells. J Cell Physiol. 228:1314–1322. 2013. View Article : Google Scholar

69 

Ghosh-Choudhury T, Mandal CC, Woodruff K, St Clair P, Fernandes G, Choudhury GG and Ghosh-Choudhury N: Fish oil targets PTEN to regulate NFkappaB for downregulation of anti-apoptotic genes in breast tumor growth. Breast Cancer Res Treat. 118:213–228. 2009. View Article : Google Scholar

70 

Moreira JD, Knorr L, Thomazi AP, Simão F, Battú C, Oses JP, Gottfried C, Wofchuk S, Salbego C, Souza DO, et al: Dietary omega-3 fatty acids attenuate cellular damage after a hippocampal ischemic insult in adult rats. J Nutr Biochem. 21:351–356. 2010. View Article : Google Scholar

71 

Yu H, Deng J and Zuo Z: High-fat diet reduces neuroprotection of isoflurane post-treatment: Role of carboxyl-terminal modulator protein-Akt signaling. Obesity (Silver Spring). 22:2396–2405. 2014. View Article : Google Scholar

72 

Liu H, Zang C, Emde A, Planas-Silva MD, Rosche M, Kühnl A, Schulz CO, Elstner E, Possinger K and Eucker J: Anti-tumor effect of honokiol alone and in combination with other anticancer agents in breast cancer. Eur J Pharmacol. 591:43–51. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Xian YF, Ip SP, Mao QQ, Su ZR, Chen JN, Lai XP and Lin ZX: Honokiol improves learning and memory impairments induced by scopolamine in mice. Eur J Pharmacol. 760:88–95. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Wong TF, Takeda T, Li B, Tsuiji K, Kitamura M, Kondo A and Yaegashi N: Curcumin disrupts uterine leiomyosarcoma cells through AKT-mTOR pathway inhibition. Gynecol Oncol. 122:141–148. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Cao F, Liu T, Xu Y, Xu D and Feng S: Curcumin inhibits cell proliferation and promotes apoptosis in human osteoclastoma cell through MMP-9, NF-κB and JNK signaling pathways. Int J Clin Exp Pathol. 8:6037–6045. 2015.

76 

Hoppe JB, Coradini K, Frozza RL, Oliveira CM, Meneghetti AB, Bernardi A, Pires ES, Beck RC and Salbego CG: Free and nano-encapsulated curcumin suppress β-amyloid-induced cognitive impairments in rats: Involvement of BDNF and Akt/GSK-3β signaling pathway. Neurobiol Learn Mem. 106:134–144. 2013. View Article : Google Scholar : PubMed/NCBI

77 

Yoshida H, Okumura N, Kitagishi Y, Nishimura Y and Matsuda S: Ethanol extract of Rosemary repressed PTEN expression in K562 culture cells. Int J appl Biol Pharm Technol. 2:316–322. 2011.

78 

Choi SY, Kim YO, Son D, Lee J, Kim S, Kim H, Kim S and Hur J: 3-[2-(3,5-Dimethoxyphenyl)vinyl]furan protects hippocampal neurons against ischemic damage. Brain Res. 1472:32–37. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Tedeschi PM, Kathari YK, Farooqi IN and Bertino JR: Leucovorin rescue allows effective high-dose pralatrexate treatment and an increase in therapeutic index in mesothelioma xenografts. Cancer Chemother Pharmacol. 74:1029–1032. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 49 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Matsuda S, Ichimura M, Ogino M, Nakano N, Minami A, Murai T and Kitagishi Y: Effective PI3K modulators for improved therapy against malignant tumors and for neuroprotection of brain damage after tumor therapy (Review). Int J Oncol 49: 1785-1790, 2016
APA
Matsuda, S., Ichimura, M., Ogino, M., Nakano, N., Minami, A., Murai, T., & Kitagishi, Y. (2016). Effective PI3K modulators for improved therapy against malignant tumors and for neuroprotection of brain damage after tumor therapy (Review). International Journal of Oncology, 49, 1785-1790. https://doi.org/10.3892/ijo.2016.3710
MLA
Matsuda, S., Ichimura, M., Ogino, M., Nakano, N., Minami, A., Murai, T., Kitagishi, Y."Effective PI3K modulators for improved therapy against malignant tumors and for neuroprotection of brain damage after tumor therapy (Review)". International Journal of Oncology 49.5 (2016): 1785-1790.
Chicago
Matsuda, S., Ichimura, M., Ogino, M., Nakano, N., Minami, A., Murai, T., Kitagishi, Y."Effective PI3K modulators for improved therapy against malignant tumors and for neuroprotection of brain damage after tumor therapy (Review)". International Journal of Oncology 49, no. 5 (2016): 1785-1790. https://doi.org/10.3892/ijo.2016.3710