Next Article in Journal
Polyketide Synthases in the Microbiome of the Marine Sponge Plakortis halichondrioides: A Metagenomic Update
Next Article in Special Issue
Programmed Cell Death Induced by (−)-8,9-Dehydroneopeltolide in Human Promyelocytic Leukemia HL-60 Cells under Energy Stress Conditions
Previous Article in Journal
Resolvin D1, a Metabolite of Omega-3 Polyunsaturated Fatty Acid, Decreases Post-Myocardial Infarct Depression
Previous Article in Special Issue
Anticancer Effects of Different Seaweeds on Human Colon and Breast Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Reactive Oxygen Species and Autophagy Modulation in Non-Marine Drugs and Marine Drugs

1
Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore 54000, Pakistan
2
Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
3
Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
4
Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
5
Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
6
Department of Radiation Oncology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
7
Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
8
Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
9
Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
10
Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
*
Author to whom correspondence should be addressed.
Mar. Drugs 2014, 12(11), 5408-5424; https://doi.org/10.3390/md12115408
Submission received: 4 September 2014 / Revised: 6 November 2014 / Accepted: 7 November 2014 / Published: 13 November 2014
(This article belongs to the Collection Marine Compounds and Cancer)

Abstract

:
It is becoming more understandable that an existing challenge for translational research is the development of pharmaceuticals that appropriately target reactive oxygen species (ROS)-mediated molecular networks in cancer cells. In line with this approach, there is an overwhelmingly increasing list of many non-marine drugs and marine drugs reported to be involved in inhibiting and suppressing cancer progression through ROS-mediated cell death. In this review, we describe the strategy of oxidative stress-based therapy and connect the ROS modulating effect to the regulation of apoptosis and autophagy. Finally, we focus on exploring the function and mechanism of cancer therapy by the autophagy modulators including inhibitors and inducers from non-marine drugs and marine drugs.

1. Introduction

1.1. Strategy of Oxidative Stress-Based Therapy

Reactive oxygen species (ROS) are essential to regulate normal cellular processes. When excess ROS stimulation appear, it may trigger DNA repair responses in normal cells to remove the ROS-mediated DNA damage [1]. For highly active metabolism, cancer cells commonly have higher levels of ROS than normal cells [2], leading to carcinogenesis by oxidative DNA damage [3] and DNA repair impairment [1]. This nature of high ROS level in cancer cells also provides a chance for drug therapy to generate overloading ROS level and induce oxidative stress-induced cell death [2,4]. Therefore, the modulation of oxidative stress is a potential strategy to anticancer therapies [5].

2. Connection between ROS and Apoptosis in Marine Drugs

In this section, we described the protective function of ROS scavengers and apoptosis induction of ROS generating drugs of marine sources as follows:

2.1. Protective Function of ROS Scavengers of Marine Sources

Several marine natural products have proved to have an anti-oxidative effect [6]. For example, aqueous extracts of the edible Gracilaria tenuistipitata have demonstrated to protect against H2O2-induced plasmid and cellular DNA damage and reverted the H2O2-induced cytotoxicity of H1299 lung cancer cells [7]. Similarly, the brown alga Sargassum horneri-derived polysaccharides reportedly exert the protective effects against H2O2-induced injury in macrophage RAW264.7 cells. The results revealed that these biological effects were achieved by downregulating intracellular ROS, nitrogen oxide, and malonic dialdehyde (MDA) levels and by upregulating the level of antioxidant system (MnSOD and GSH-Px) in RAW264.7 cells [8]. Similarly, a lipid-soluble pigment of marine carotenoid astaxanthin can inhibit lipopolysaccaride-induced ROS generation and cytotoxicity via upregulation of superoxide dismutase (SOD) and catalase in mononuclear U937 cells [9].

2.2. Apoptosis Induction of ROS Generating Drugs of Marine Sources

In contrast, the accumulating evidence shows that several marine-derived extracts and compounds have the ROS inducible effects on different cancer cell lines. For example, 10-acetylirciformonin B (10AB), a marine sponge furanoterpenoid derived from irciformonin B [10], was reported to induce apoptosis via ROS generation in different cancer cell lines. Pretreatment of a ROS scavenger N-acetyl-l-cysteine to leukemia HL 60 cells drastically impaired 10AB-induced apoptosis, supporting that ROS generation was involved in irciformonin B-induced cytotoxicity of leukemia cells. Importantly, the protein expressions of Bcl-xL and Bcl-2, and caspase inhibitors (XIAP and surviving) were considerably repressed and the pro-apoptotic protein Bax was increased in 10AB treated leukemia HL 60 cells [11]. For another marine sponge derived compound Fascaplysin, it was apoptosis inducible in a chemoresistant NCI-H417 SCLC cells through ROS generation. Moreover, it was noted that fascaplysin worked synergistically with topoisomerase I-directed camptothecin and 10-hydroxy-camptothecin [12].
Dicitrinone B, a marine fungal metabolite, reportedly induced apoptosis via ROS generation in human malignant melanoma A375 cells. After pan-caspase inhibitor treatment to A375 cells, the dicitrinone B-induced ROS generation and apoptosis was abolished [13], suggesting that caspase pathway was involved in its ROS generation and apoptosis effects. A 48 kDa glycoprotein, isolated from a marine macroalga Codium decorticatum, was reported to induce ROS and apoptosis in breast cancer MDA-MB-231 cells through the intrinsic apoptosis pathway [14]. Surprisingly, it has been shown that lamellarin D, a marine alkaloid isolated from a marine mollusk Lamellaria sp. [15] and various ascidians [16], can induce ROS-mediated senescence in the absence of functional mitochondria in mouse leukemia P388 cells [17]. Marine triprenyl toluquinones and toluhydroquinones, originally purified from the Arminacean nudibranch Leminda millecra, have a similar ROS inducible effect to esophageal cancer WHCO1 cells [18]. Additionally, both methanolic extracts [19] and ethanolic extracts [20] of the editable red alga Gracilaria tenuistipitata showed the ROS generation and apoptosis induction in oral cancer cells. Ethyl acetate extracts from three marine algae (Colpomenia sinuosa, Halimeda discoidae, and Galaxaura oblongata) also displayed a ROS-mediated antiproliferative effect against human liver cancer and leukemia cells [21].
It had been reviewed that different natural products may induce different degrees of apoptosis and autophagy depending on their ROS modulating effect [22]. The marine drugs mentioned above have shown a ROS-mediated apoptotic effect, however, the possible roles of autophagy in these mechanisms warrant for further investigation.

3. Brief Introduction of Autophagy and Connection between ROS and Autophagy

In this section, we briefly introduce the autophagy and describe the relationship between ROS and autophagy as follows:

3.1. Brief Introduction of Autophagy

Autophagy is a “self-eating” behavior to ship cellular proteins and damaged organelles to lysosomes for recycling and it subsequently maintains the energy balance for cell survival during cell stress or starvation [23]. However, autophagy also reviewed to induce cell death in some cases [24]. There is a tremendously increasing amount of information regarding biology of autophagy. A substantial fraction of knowledge has been added into different steps of autophagy and it is now known that it is a highly regulated, multi-step molecular mechanism that initializes with induction, autophagosome nucleation, expansion and completion. Later steps of autophagy include lysosome fusion, degradation and recycling [25].
Structural studies have provided near complete resolution of protein network of mechanism of autophagy and mounting evidence suggested that initialization occurred through activation of AuTophaGy related 1 (Atg1) complex. It is multi-component machinery formed by assembly of Atg1, Atg13 and Atg17. Atg1 is a kinase that needs association of Atg13 and Atg17 for its activation. Vesicle nucleation is the subsequent process triggered by activation of the Vps34 and Beclin-1/Atg6. Autophagosome formation requires recruitment of proteins and lipids. Atg7 (E1-like) and Atg3 (E2-like) modulate vesicle elongation and completion by conjugation of phosphatidylethanolamine to microtubule-associated protein1 light chain 3 (LC3)/Atg8 which is initially processed by Atg4 [26]. Among them, the key step in autophagy is the proteolytic cleavage of LC3 to form LC3-I and subsequently modified to from LC3-II [27]. Moreover, Atg7 and Atg10 can join together to modulate the interaction between Atg12 and Atg5, and they are finally transferred to Atg16.

3.2. ROS May Lead to Autophagy

ROS are essential in maintaining normal cellular physiology, but ROS dysregulation may lead to tumor development and progression. Mitochondrial ROS generation play an important role for apoptosis and autophagy [25]. The autophagy may be induced to survival and cell death pathways in response to cellular oxidative stress [24]. Therefore, some of ROS-inducible drugs, such as 2-methoxyestradiol and arsenic trioxide, are used for cancer treatments [28]. Similarly, reduced scytonemin isolated from a terrestrial benthic cyanobacterium, Nostoc commune, induces ROS-based autophagy in human T-lymphoid Jurkat cells [29]. In the next sections, we will summarize many autophagy inhibitors and inducers derived from non-marine drugs and marine drugs to discuss the cancer therapy of those autophagy modulators.

4. Autophagy Inhibitors and Inducers from Non-Marine Drugs in Cancer Therapy

Accumulating evidence showed that it was inter-compensatory between autophagy and apoptosis. For example, autophagy may have a cytotoxic role [30]. When the autophagy was induced, the cell death was promoted. For example, autophagic degradation of protein phosphatase Fap-1 was reported to enhance Fas-induced apoptosis. When cells displayed high autophagy, p62 recruited more Fap-1 for degradation and functional Fas ligands and receptors were highly maintained to activate more apoptotic signaling [31,32]. In this section, we described the function of the non-marine drugs derived autophagy inhibitors, autophagy inducers, clinical trial of autophagy inhibitor, clinical trial of autophagy inducers, and established anticancer drugs combined with autophagy inhibitors as follows:

4.1. Autophagy Inhibitors

Autophagy may have a cytoprotective role [30]. When the autophagy was inhibited, the cell death was promoted. For example, an autophagy inhibitor 3-methyladenine (3-MA) was reported to increase the apoptosis inducing potential of breast cancer MDA-MB 231 cells treated with a commercial mixture of tocotrienols and tocopherols (Tocomin®), which were isolated from palm oil/palm fruits [33,34]. It was revealed that mixture of tocotrienols and tocopherols can inhibit phosphoinositide 3-kinase (PI3K) and mammalian target of rapamycin (serine/threonine kinase) (mTOR) pathways, and induce the cytoprotective autophagic response in MDA-MB 231 cells, which could be overcome through inhibition of autophagy [34].

4.2. Autophagy Inducers

In accordance with the notion that Akt-mTOR signaling is a negative regulator of autophagy [35], gambogic acid, isolated from gamboge resin, can enhance the ROS accumulation and suppress phosphorylation of both Akt (S473) and mTOR (S2448) to induce autophagy in colorectal cancer HCT116 cells [36]. It is relevant to mention that extracellular signal-regulated kinases (ERK) pathway is also involved in initiation of autophagic response in hepatocellular carcinoma (HCC) cells as well as in mice xenografted with HCC cells [37]. A histone deacetylase inhibitor (HDACi) MGCD0103 has been shown to inhibit autophagy by functionalizing PI3K/AKT/mTOR pathway as well as caspases in B-cell chronic lymphocytic leukemia cells (CLL) [38]. Consistently, ATP-competitive mTOR kinase inhibitors (CC214-1 and CC214-2) were effective against rapamycin-resistant mTORC1 signaling to induce autophagy and prevent tumor cell death [39]. Cathepsin S, a lysosomal cysteine protease, was reported to overexpress in glioblastoma cells [40]. Inhibition of cathepsin S by its inhibitor Z-FL-COCHO (ZFL) can induce autophagy and mitochondrial-based apoptosis in glioblastoma cells. In autophagy-inhibitory glioblastoma cells by treating an autophagy inhibitor 3-MA or Beclin-1 shRNA, cathepsin S inhibition-induced apoptosis were drastically reduced. In cathepsin S-inhibitory glioblastoma cells, ROS-mediated PI3K/AKT/mTOR/p70S6K signaling pathway was inhibited and c-Jun N-terminal kinase (JNK) was activated [41].

4.3. Clinical Trial of Autophagy Inhibitors

Hydoxychloroquine (HCQ), a drug derived from quinolone, is antiproliferative to human dermal fibroblasts and induces autophagy in terms of upregulation of Beclin-1 [42,43]. Metastatic pancreatic cancer patients previously treated with HCQ at a dosage of 400 mg or 600 mg twice daily did not show considerable autophagy inhibition or therapeutic value [44]. Recently, the combined treatments of autophagy inhibitor HCQ with some drugs are being tested in preclinical and ongoing clinical cancer studies [45]. For example, HCQ is noted to effectively inhibit cancer growth in combination with epirubicin in xenografted mice [46]. However, the dosages of HCQ applied to inhibit autophagy are inconsistently functional in clinic studies [47].
Additionally, Lys05, a water-soluble salt of the lead compound Lys01 show that Lys05 targets to impair autophagy and inhibit tumor growth without toxicity under lower doses of Lys05 in mice studies [47]. These results suggest that Lys05 is warranted for further clinical trial in future.

4.4. Established Anticancer Drugs Combined with Autophagy Inhibitors

Emerging evidence has shed light on the fact that autophagy induced resistance against chemotherapeutic drugs in cancer cells, i.e., a cytoprotective role of autophagy. In the following section we will discuss accumulating in vitro and in vivo evidence to understand how autophagy inhibition can be helpful in maximizing chemotherapeutic drug induced therapeutic effects in cancer cells. For example, treating with 3-MA or Beclin-1 siRNA to inhibit autophagy in colorectal cancer HCT116 and RKO cells, the low dose (20–50 nM) of a clinical drug for topoisomerase I inhibitor camptothecin-induced senescence was turned to caspase 3-dependent apoptosis [48]. For the combined treatment of clinical drugs sorafenib and vorinostat (the multikinase and HDAC inhibitors, respectively), its growth inhibitory efficacy can be enhanced in the autophagy inhibitor 3-MA treated hepatoma cells [49]. Inhibition of autophagy by beclin1 siRNA in ovarian cancer SKOV3/DDP cells has been noted to considerably increase cisplatin-induced apoptosis [50]. By pre-treatment of chloroquine for autophagy inhibition, DNA damaging agent 5-fluorouracil-induced cell death were remarkably increased in gallbladder carcinoma SGC-996 and GBC-SD cells [51]. Similarly, inhibition of autophagy by chloroquine can restore sensitivity of resistant lung cancer H3122CR-1 cells to crizonitib (PF02341066, the inhibitor of ALK fusion oncoprotein) [52].
Similar cytoprotective role of autophagy was also reported in literature. For example, overexpressing high-mobility group nucleosome-binding domain 5 (HMGN5) in osteosarcoma U2OS and MG63 cell lines can induce resistance against chemotherapeutic drugs such as doxorubicin, cisplatin, and methotrexate via inducing autophagy [53]. Inhibition of autophagy with clomipramine or metformin can enhance apoptosis and show the cytoprotective role of autophagy. Gene silencing with AMP-dependent protein kinase (AMPK) siRNA can substantially inhibit AMPK-induced downstream autophagy signaling and induce apoptosis in clinical trial drug enzalutamide (ENZA) treated prostate cancer cells. In mice orthotopically transplanted with ENZA-resistant cells, the combined treatment of ENZA and autophagy inhibitors (clomipramine and metformin) can reduce tumor growth compared to control groups [54]. The signal transducer and activator of transcription 3 (STAT3) was activated by oxidative stress. Downregulated STAT3 in pancreatic cancer cells also reported to inhibit cell growth through repressing autophagy induced by the treatment of Nexrutine(R) (Nx), a bark extract from Phellodendron amurense [55].
There is an exciting piece of evidence highlighting diametrically opposed role of autophagy as a pro-survival (cytoprotective), as well as a cell death-inducing (cytotoxic) role in cancer cells. For the example of cytotoxic role of autophagy, detailed investigation revealed that Akt activation and autophagy inhibition were responsible to the acquired resistance to sorafenib. A novel ATP-competitive pan-Akt inhibitor GDC0068 can reverse the acquired resistance to sorafenib, the first-line clinical drug for advanced HCC and autophagy was activated to be cytotoxic [56]. Similar cytotoxic role of autophagy was also reported that enforced expression of an imprinted tumor suppressor gene GTP-binding RAS-like 3 (DIRAS3 or ARHI) in DIRAS3-deficient ovarian cancer cells may induce autophagy and tumor dormancy [57]. In cells reconstituted with DIRAS3, growth factor-mediated intracellular signaling through PI3K and Ras/MAP kinase pathways were inhibited. Additionally, DIRAS3 can downregulate PI3K/AKT and Ras/ERK pathway and reduce phosphorylation of forkhead box O3 (FOXO3a) that facilitated transportation of FOXO3a to induce expression of autophagy-related genes (ATG4, MAP-LC3-I and Rab7) for maturation of autophagosomes and fusion with lysosomes [57]. Furthermore, DIRAS3 was reported to trigger assembly of autophagosome initiation complex to induce autophagy in dormant, nutrient-deprived ovarian cancer cells [58].

5. Autophagy Inhibitors and Inducers from Marine Drugs in Cancer Therapy

As shown in Table 1, in this section we described the autophagy inhibitors and inducers of marine drugs of several species of the marine sponges, algae, bacteria/fungi/cyanobacteria, and other marine-derived compounds as follows:
Table 1. A list of bioactive ingredients that act as autophagy inhibitors and inducers.
Table 1. A list of bioactive ingredients that act as autophagy inhibitors and inducers.
FunctionMarine SourceSourceChemicalTargetReferences
Autophagy inhibitorsMarine SpongePetrosaspongia nigraPetrosaspongiolide MBeclin-1 ↓[59,60] *
Marine bacteriumStreptomyces spp.BafilomycinsLC3-II ↓[61]
Autophagy inducersMarine SpongeHaliclona sp.Manzamine ALC3-II ↑
P62/SQTM1↑
[62]
[63] *
Haliclona sp.PapuamineLC3-II ↑[64,65] *
Cliona celataClionamines A–DLC3 ↑[66]
Geodia japonicaStellettin ALC3-II ↑[67,68] *
Rhabdastrella globostellataRhabdastrellic acid-ApAkt ↓[69,70] *
AlgaGreen algae
(Enteromorpha intestinalis;
Rhizoclonium riparium)
Methanolic extractsLC3-II ↑[71]
Red alga
(Laurencia dendroidea)
Sesquiterpene elatolendoplasmic reticulum extension ↑[72]
[73] *
Brown algaeFucoxanthinLC3-II ↑
Beclin-1 ↑
[74]
[75] *
Marine bacterium/fungus/cyanobacteriumSalinispora tropica; Salinispora arenicolaSalinosporamide AATG5 ↑
ATG7 ↑
[76]
[77] *
Chondrostereum spHirsutanolLC3-II ↑
ROS↑
[78]
[79] *
Penicillium communeSD118-xanthocillin X (1)LC3-II ↑
mTOR, ERK ↓
[80]
Leptolyngbya sp.CoibamideLC3-II ↑[81]
* References state that autophagy-modulating drugs also have an apoptosis modulating effect.

5.1. Marine Sponge

5.1.1. Autophagy Inhibitors

5.1.1.1. Petrosaspongia Nigra

Petrosaspongiolide M, a γ-hydroxybutenolide terpenoid isolated from a marine sponge Petrosaspongia nigra [82], can exert inhibitory effects on autophagy in human macrophage U937 cells in terms of downregulation of Beclin-1 level [59].

5.1.2. Autophagy Inducers

5.1.2.1. Haliclona sp.

Manzamine A, a kind of alkaloids for the uncoupler of vacuolar ATPases isolated from a marine sponge Haliclona, was reported to be a potential autophagy inducer. Mechanistically, manzamine A exerted its effects via increasing LC3-II and p62/SQSTM1 in pancreatic cancer cells [62]. Moreover, manzamine A can resensitize TRAIL-induced apoptosis in the pancreatic cancer AsPC-1 cells [63].
Papuamine, one of the isolated compounds from Haliclona sp. has been noted to decrease survival of breast cancer MCF-7 cells. Papuamine treated MCF-7 cells revealed an increase in expression of LC3 after 4 h treatment. Overall it suggested that papuamine induced early autophagy in MCF-7 cells that later activated JNK [64].

5.1.2.2. Cliona celata

Aminosteroids clionamines A–D, isolated from South African sponge Cliona celata, was reported to induce autophagosome accumulation in terms of formation of cytoplasmic punctate Green Fluorescent Protein (GFP)-LC3 [66]. Clionamine B (2) was also reported to induce autophagy in human breast cancer MCF-7 cells [83].

5.1.2.3. Geodia japonica

Stellettin, isolated from a marine sponge Geodia japonica, has been shown to induce autophagy in B16F10 murine melanoma cells. Increased LC3-II and its co-localization with tyrosinase indicated removal of deglycosylated and unfolded proteins [67].

5.1.2.4. Rhabdastrella globostellata

Rhabdastrellic acid-A, an isomalabaricane Triterpenoid purified from a marine sponge Rhabdastrella globostellata, also notably induced autophagy in human lung cancer A549 cells. In Atg5 knockdown cells, rhabdastrellic acid-A mediated autophagy was impaired. pAkt was reduced in rhabdastrellic acid-A treated A549 cells and interestingly, transfecting constitutively active Akt in A549 cells can inhibit rhabdastrellic acid-A induced autophagy [69].

5.2. Alga

5.2.1. Autophagy Inducers

5.2.1.1. Enteromorpha intestinalis and Rhizoclonium riparium

Algal methanolic extracts from green alga Enteromorpha intestinalis and Rhizoclonium riparium, the saline/brackish water algae from Sundarbans, can induce autophagy in HeLa cells as evidenced by considerably enhanced expression of LC3-II [71].

5.2.2. Laurencia dendroidea

Sesquiterpene elatol, the major bioactive compound of red seaweed Laurencia dendroidea, was reported to be an antiproliferative agent against Leishmania amazonensis with endoplasmic reticulum extension, which is an autophagy marker [72].

5.2.3. Brown Algae

Fucoxanthin, a major carotenoid found in edible brown algae, was reported to be dose-responsively cytotoxic and G0/G1 arrest of HeLa cells without apoptosis change. Alternatively, autophagy-based cytotoxicity of fucoxanthin-treated HeLa cells was found involving the inhibition of Akt/mTOR signaling pathway [74].

5.3. Marine Bacterium/Fungus/Cyanobacterium

5.3.1. Autophagy Inhibitors

5.3.1.1. Streptomyces spp.

Eight bafilomycins (A1, B1, D, F, G, H, I, and J), purified from Streptomyces spp. of marine habitats, were proved to be potent inhibitors of autophagy in terms of automated microscopy screening assay-based punctate formation of EGFP-LC3 (autophagosome accumulation) and the Western blot-based EGFP-LC3 degradation assay [61]. Proteinase inhibitors, such as clasto-lactacystinblactone (LA) or epoxomicin (Epo) were recently reported to induce autophagy through inhibition of PI3K-Akt-mTOR pathway in human retinal pigment epithelial ARPE-19 cells [84]. Using the autophagy inhibitor bafilomycin A1, the protective effects of LA or Epo against menadione-induced oxidative injuries in ARPE-19 cells were reverted.

5.3.2. Autophagy Inducers

5.3.2.1. Salinispora tropica and Salinispora arenicola

Salinosporamide A, a potent proteasome inhibitor from marine bacteria Salinispora tropica and Salinispora arenicola, was reported to induce autophagy through a phospho-eukaryotic translation initiation factor 2α (eIF2α) pathway to reduce proteotoxic stresses in human prostate cancer cells [76].

5.3.2.2. Chondrostereum sp.

Hirsutanol is a sesquiterpene isolated from marine fungus Chondrostereum sp. in the coral Sarcophyton tortuosum [85]. In hirsutanol-treated breast cancer MCF-7 cells, LC3-I to LC3-II conversion and ROS induction were markedly increased as evidenced by Western blot assay and flow cytometry [78].

5.3.2.3. Penicillium commune

SD118-xanthocillin X, isolated from the marine fungus Penicillium commune, can induce autophagy in hepatocellular carcinoma HepG2 cells. There was a conversion of LC3-I to LC3-II, following lipidation as it incorporates into the nascent membrane of the autophagosome. Mechanistically it was noted that SD118-xanthocillin regulated different modulators of autophagy. It exerted its autophagy inducing effects via inhibition of phosphorylation of mTOR and ERK1/2. Additionally, Bcl-2 mediated inhibition of Beclin-1 to suppress autophagy was also attenuated via inhibition of Bcl-2 by SD118-xanthocillin [80].

5.3.2.4. Leptolyngbya sp.

Coibamide A, a depsipeptide derived from marine cyanobacterium Leptolyngbya sp., showed a cytotoxicity in the dose-responsive and time-dependent manner in human glioblastoma cells and mouse embryonic fibroblasts (MEF) [81]. In coibamide A treated human glioblastoma U87-MG cells, LC3-II expression was notably increased. Coibamide A also induced the autophagosome accumulation in glioblastoma and MEF cells. Detailed mechanistic insights indicated that accumulation of autophagosomes was independent of mTOR-mediated signaling.

5.4. Other Marine-Derived Agents

Marine-derived agents, including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are also potent inducers of autophagy as indicated by formation of autophagosomes in DHA- or EPA-treated lung adenocarcinoma A549 cells [86].
The Na+/K+-ATPases (NKA) inhibitor cardiac glycosides, a family of natural or synthetic steroid hormones isolated from marine or terrestrial natural products [87], can exert their potent anti-cancer properties via activation of Src in the upstream of MEK1/2 and ERK1/2 pathway in human non-small cell lung cancer A549 and H460 cells [88]. Src inhibition by its inhibitor PP2 or siRNA can remarkably repress cardiac glycosides-induced MEK1/2 and ERK1/2 phosphorylation and autophagic cell death. Moreover, ROS was also noted to be accumulated and contributed to cardiac glycosides-induced Src mediated autophagic response in lung cancer cells.

6. Conclusions

In this review, we summarized how ROS-mediated molecular networks may result in autophagy. The autophagic effects of both clinical drugs and natural products-derived extracts and pure compounds were discussed. In the example of many autophagy modulators (inducers and inhibitors) from non-marine drugs and marine drugs, ROS changes and signaling was demonstrated to be involved in autophagy. Many marine drugs with autophagy were also summarized from marine sponges, alga, and marine bacteria/fungi/ cyanobacteria. It suggests that marine drugs with ROS modulating effect have a potential to modulate the autophagy of cancer cells to improve cancer therapy.

Acknowledgments

This work was partly supported by funds of the Ministry of Science and Technology (MOST 103-2320-B-037-008), the Kaohsiung Medical University “Aim for the Top Universities Grant, grant No. KMU-TP103A33”, the National Sun Yat-sen University-KMU Joint Research Project (#NSYSU-KMU 103-p014), and the Health and welfare surcharge of tobacco products, the Ministry of Health and Welfare, Taiwan, Republic of China (MOHW103-TD-B-111-05).

Author Contributions

A.-A.F., J.-Y.T., and H.-W.C. integrated different points of searched literatures, and drafted the manuscript. S.F., M.-F.H. and K.-T.L. conceived the idea, did literature search on specific points, and involved in discussion. All authors read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Trachootham, D.; Alexandre, J.; Huang, P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach? Nat. Rev. Drug Discov. 2009, 8, 579–591. [Google Scholar] [CrossRef]
  2. Nogueira, V.; Hay, N. Molecular pathways: Reactive oxygen species homeostasis in cancer cells and implications for cancer therapy. Clin. Cancer Res. 2013, 19, 4309–4314. [Google Scholar] [CrossRef] [PubMed]
  3. Montero, A.J.; Jassem, J. Cellular redox pathways as a therapeutic target in the treatment of cancer. Drugs 2011, 71, 1385–1396. [Google Scholar] [CrossRef] [PubMed]
  4. Ivanova, D.; Bakalova, R.; Lazarova, D.; Gadjeva, V.; Zhelev, Z. The impact of reactive oxygen species on anticancer therapeutic strategies. Adv. Clin. Exp. Med. 2013, 22, 899–908. [Google Scholar] [PubMed]
  5. Gorrini, C.; Harris, I.S.; Mak, T.W. Modulation of oxidative stress as an anticancer strategy. Nat Rev. Drug Discov. 2013, 12, 931–947. [Google Scholar] [CrossRef] [PubMed]
  6. Lee, J.C.; Hou, M.F.; Huang, H.W.; Chang, F.R.; Yeh, C.C.; Tang, J.Y.; Chang, H.W. Marine algal natural products with anti-oxidative, anti-inflammatory, and anti-cancer properties. Cancer Cell Int. 2013, 13, 55. [Google Scholar] [CrossRef] [PubMed]
  7. Yang, J.I.; Yeh, C.C.; Lee, J.C.; Yi, S.C.; Huang, H.W.; Tseng, C.N.; Chang, H.W. Aqueous extracts of the edible Gracilaria tenuistipitata are protective against H2O2-induced DNA damage, growth inhibition, and cell cycle arrest. Molecules 2012, 17, 7241–7254. [Google Scholar] [CrossRef] [PubMed]
  8. Wen, Z.S.; Liu, L.J.; OuYang, X.K.; Qu, Y.L.; Chen, Y.; Ding, G.F. Protective effect of polysaccharides from Sargassum horneri against oxidative stress in RAW264.7 cells. Int. J. Biol. Macromol. 2014, 68C, 98–106. [Google Scholar] [CrossRef] [PubMed]
  9. Franceschelli, S.; Pesce, M.; Ferrone, A.; De Lutiis, M.A.; Patruno, A.; Grilli, A.; Felaco, M.; Speranza, L. Astaxanthin treatment confers protection against oxidative stress in U937 cells stimulated with lipopolysaccharide reducing O2- production. PLoS One 2014, 9, e88359. [Google Scholar] [CrossRef] [PubMed]
  10. Su, J.H.; Chang, W.B.; Chen, H.M.; El-Shazly, M.; Du, Y.C.; Kung, T.H.; Chen, Y.C.; Sung, P.J.; Ho, Y.S.; Kuo, F.W.; et al. 10-acetylirciformonin B, a sponge furanoterpenoid, induces DNA damage and apoptosis in leukemia cells. Molecules 2012, 17, 11839–11848. [Google Scholar] [CrossRef] [PubMed]
  11. Shih, H.C.; El-Shazly, M.; Juan, Y.S.; Chang, C.Y.; Su, J.H.; Chen, Y.C.; Shih, S.P.; Chen, H.M.; Wu, Y.C.; Lu, M.C. Cracking the cytotoxicity code: Apoptotic induction of 10-acetylirciformonin B is mediated through ROS generation and mitochondrial dysfunction. Mar. Drugs 2014, 12, 3072–3090. [Google Scholar] [CrossRef] [PubMed]
  12. Hamilton, G. Cytotoxic effects of fascaplysin against small cell lung cancer cell lines. Mar. Drugs 2014, 12, 1377–1389. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, L.; Gong, M.W.; Peng, Z.F.; Zhou, T.; Ying, M.G.; Zheng, Q.H.; Liu, Q.Y.; Zhang, Q.Q. The marine fungal metabolite, dicitrinone B, induces A375 cell apoptosis through the ROS-related caspase pathway. Mar. Drugs 2014, 12, 1939–1958. [Google Scholar] [CrossRef] [PubMed]
  14. Thangam, R.; Senthilkumar, D.; Suresh, V.; Sathuvan, M.; Sivasubramanian, S.; Pazhanichamy, K.; Gorlagunta, P.K.; Kannan, S.; Gunasekaran, P.; Rengasamy, R.; et al. Induction of ROS-dependent mitochondria-mediated intrinsic apoptosis in MDA-MB-231 cells by glycoprotein from Codium decorticatum. J. Agric. Food Chem. 2014, 62, 3410–3421. [Google Scholar] [CrossRef] [PubMed]
  15. Andersen, R.J.; Faulkner, D.J.; He, C.H.; Van Duyne, G.D.; Clardy, J. Metabolites of the marine prosobranch mollusk Lamellaria sp. J. Am. Chem. Soc. 1985, 107, 5492–5495. [Google Scholar] [CrossRef]
  16. Davis, R.A.; Carroll, A.R.; Pierens, G.K.; Quinn, R.J. New lamellarin alkaloids from the Australian ascidian, Didemnum chartaceum. J. Nat. Prod. 1999, 62, 419–424. [Google Scholar] [CrossRef] [PubMed]
  17. Ballot, C.; Martoriati, A.; Jendoubi, M.; Buche, S.; Formstecher, P.; Mortier, L.; Kluza, J.; Marchetti, P. Another facet to the anticancer response to lamellarin D: Induction of cellular senescence through inhibition of topoisomerase I and intracellular ROS production. Mar. Drugs 2014, 12, 779–798. [Google Scholar] [CrossRef] [PubMed]
  18. Whibley, C.E.; McPhail, K.L.; Keyzers, R.A.; Maritz, M.F.; Leaner, V.D.; Birrer, M.J.; Davies-Coleman, M.T.; Hendricks, D.T. Reactive oxygen species mediated apoptosis of esophageal cancer cells induced by marine triprenyl toluquinones and toluhydroquinones. Mol. Cancer Ther. 2007, 6, 2535–2543. [Google Scholar] [CrossRef] [PubMed]
  19. Yeh, C.C.; Yang, J.I.; Lee, J.C.; Tseng, C.N.; Chan, Y.C.; Hseu, Y.C.; Tang, J.Y.; Chuang, L.Y.; Huang, H.W.; Chang, F.R.; et al. Anti-proliferative effect of methanolic extract of Gracilaria tenuistipitata on oral cancer cells involves apoptosis, DNA damage, and oxidative stress. BMC Complement Altern. Med. 2012, 12, 142. [Google Scholar] [CrossRef] [PubMed]
  20. Yeh, C.C.; Tseng, C.N.; Yang, J.I.; Huang, H.W.; Fang, Y.; Tang, J.Y.; Chang, F.R.; Chang, H.W. Antiproliferation and induction of apoptosis in Ca9-22 oral cancer cells by ethanolic extract of Gracilaria tenuistipitata. Molecules 2012, 17, 10916–10927. [Google Scholar] [CrossRef] [PubMed]
  21. Huang, H.L.; Wu, S.L.; Liao, H.F.; Jiang, C.M.; Huang, R.L.; Chen, Y.Y.; Yang, Y.C.; Chen, Y.J. Induction of apoptosis by three marine algae through generation of reactive oxygen species in human leukemic cell lines. J. Agric. Food Chem. 2005, 53, 1776–1781. [Google Scholar] [CrossRef] [PubMed]
  22. Chang, H.W. The fate of marine algal natural products-treated cells depend on it ROS modulating effects. J. Rashid Latif Med. College 2013, 2, 8–10. [Google Scholar]
  23. Mizushima, N. Autophagy: Process and function. Genes Dev. 2007, 21, 2861–2873. [Google Scholar] [CrossRef] [PubMed]
  24. Li, L.; Ishdorj, G.; Gibson, S.B. Reactive oxygen species regulation of autophagy in cancer: Implications for cancer treatment. Free Radic. Biol. Med. 2012, 53, 1399–1410. [Google Scholar] [CrossRef] [PubMed]
  25. Li, Z.Y.; Yang, Y.; Ming, M.; Liu, B. Mitochondrial ROS generation for regulation of autophagic pathways in cancer. Biochem. Biophys. Res. Commun. 2011, 414, 5–8. [Google Scholar] [CrossRef] [PubMed]
  26. Denton, D.; Nicolson, S.; Kumar, S. Cell death by autophagy: Facts and apparent artefacts. Cell Death Differ. 2012, 19, 87–95. [Google Scholar] [CrossRef]
  27. Tanida, I.; Sou, Y.S.; Ezaki, J.; Minematsu-Ikeguchi, N.; Ueno, T.; Kominami, E. HsAtg4B/HsApg4B/autophagin-1 cleaves the carboxyl termini of three human Atg8 homologues and delipidates microtubule-associated protein light chain 3- and GABAA receptor-associated protein-phospholipid conjugates. J. Biol. Chem. 2004, 279, 36268–36276. [Google Scholar] [CrossRef]
  28. Azad, M.B.; Chen, Y.; Gibson, S.B. Regulation of autophagy by reactive oxygen species (ROS): Implications for cancer progression and treatment. Antioxid. Redox Signal. 2009, 11, 777–790. [Google Scholar] [CrossRef] [PubMed]
  29. Itoh, T.; Tsuzuki, R.; Tanaka, T.; Ninomiya, M.; Yamaguchi, Y.; Takenaka, H.; Ando, M.; Tsukamasa, Y.; Koketsu, M. Reduced scytonemin isolated from Nostoc commune induces autophagic cell death in human T-lymphoid cell line Jurkat cells. Food Chem. Toxicol. 2013, 60, 76–82. [Google Scholar] [CrossRef] [PubMed]
  30. Gewirtz, D.A. The four faces of autophagy: Implications for cancer therapy. Cancer Res. 2014, 74, 647–651. [Google Scholar] [CrossRef] [PubMed]
  31. Joshi, S.; Ryan, K.M. Autophagy chews Fap to promote apoptosis. Nat. Cell Biol. 2014, 16, 23–25. [Google Scholar] [CrossRef] [PubMed]
  32. Gump, J.M.; Staskiewicz, L.; Morgan, M.J.; Bamberg, A.; Riches, D.W.; Thorburn, A. Autophagy variation within a cell population determines cell fate through selective degradation of Fap-1. Nat. Cell Biol. 2014, 16, 47–54. [Google Scholar] [CrossRef] [PubMed]
  33. Yu, W.; Simmons-Menchaca, M.; Gapor, A.; Sanders, B.G.; Kline, K. Induction of apoptosis in human breast cancer cells by tocopherols and tocotrienols. Nutr. Cancer 1999, 33, 26–32. [Google Scholar] [CrossRef] [PubMed]
  34. Tran, A.T.; Ramalinga, M.; Kedir, H.; Clarke, R.; Kumar, D. Autophagy inhibitor 3-methyladenine potentiates apoptosis induced by dietary tocotrienols in breast cancer cells. Eur. J. Nutr. 2014, in press [PMID: 24830781]. [Google Scholar]
  35. Liang, C. Negative regulation of autophagy. Cell Death Differ. 2010, 17, 1807–1815. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, H.; Lei, Y.; Yuan, P.; Li, L.; Luo, C.; Gao, R.; Tian, J.; Feng, Z.; Nice, E.C.; Sun, J. ROS-mediated autophagy induced by dysregulation of lipid metabolism plays a protective role in colorectal cancer cells treated with gambogic acid. PLoS One 2014, 9, e96418. [Google Scholar] [CrossRef] [PubMed]
  37. Gong, K.; Zhang, Z.; Chen, Y.; Shu, H.B.; Li, W. Extracellular signal-regulated kinase, receptor interacting protein, and reactive oxygen species regulate shikonin-induced autophagy in human hepatocellular carcinoma. Eur. J. Pharmacol. 2014, 738, 142–152. [Google Scholar] [CrossRef] [PubMed]
  38. El-Khoury, V.; Pierson, S.; Szwarcbart, E.; Brons, N.H.; Roland, O.; Cherrier-De Wilde, S.; Plawny, L.; van Dyck, E.; Berchem, G. Disruption of autophagy by the histone deacetylase inhibitor MGCD0103 and its therapeutic implication in B-cell chronic lymphocytic leukemia. Leukemia 2014, 28, 1636–1646. [Google Scholar] [CrossRef] [PubMed]
  39. Gini, B.; Zanca, C.; Guo, D.; Matsutani, T.; Masui, K.; Ikegami, S.; Yang, H.; Nathanson, D.; Villa, G.R.; Shackelford, D.; et al. The mTOR kinase inhibitors, CC214-1 and CC214-2, preferentially block the growth of EGFRvIII-activated glioblastomas. Clin. Cancer Res. 2013, 19, 5722–5732. [Google Scholar] [CrossRef] [PubMed]
  40. Flannery, T.; McQuaid, S.; McGoohan, C.; McConnell, R.S.; McGregor, G.; Mirakhur, M.; Hamilton, P.; Diamond, J.; Cran, G.; Walker, B.; et al. Cathepsin S expression: An independent prognostic factor in glioblastoma tumours—A pilot study. Int. J. Cancer 2006, 119, 854–860. [Google Scholar] [CrossRef] [PubMed]
  41. Zhang, L.; Wang, H.; Xu, J.; Zhu, J.; Ding, K. Inhibition of cathepsin S induces autophagy and apoptosis in human glioblastoma cell lines through ROS-mediated PI3K/AKT/mTOR/p70S6K and JNK signaling pathways. Toxicol. Lett. 2014, 228, 248–259. [Google Scholar] [CrossRef] [PubMed]
  42. Oikarinen, A. Hydroxychloroquine induces autophagic cell death of human dermal fibroblasts: Implications for treating fibrotic skin diseases. J. Invest. Dermatol. 2009, 129, 2333–2335. [Google Scholar]
  43. Ramser, B.; Kokot, A.; Metze, D.; Weiss, N.; Luger, T.A.; Bohm, M. Hydroxychloroquine modulates metabolic activity and proliferation and induces autophagic cell death of human dermal fibroblasts. J. Invest. Dermatol. 2009, 129, 2419–2426. [Google Scholar] [CrossRef] [PubMed]
  44. Wolpin, B.M.; Rubinson, D.A.; Wang, X.; Chan, J.A.; Cleary, J.M.; Enzinger, P.C.; Fuchs, C.S.; McCleary, N.J.; Meyerhardt, J.A.; Ng, K.; et al. Phase II and pharmacodynamic study of autophagy inhibition using hydroxychloroquine in patients with metastatic pancreatic adenocarcinoma. Oncologist 2014, 19, 637–638. [Google Scholar] [CrossRef] [PubMed]
  45. Yang, Z.J.; Chee, C.E.; Huang, S.; Sinicrope, F.A. The role of autophagy in cancer: Therapeutic implications. Mol. Cancer Ther. 2011, 10, 1533–1541. [Google Scholar] [CrossRef] [PubMed]
  46. Chittaranjan, S.; Bortnik, S.; Dragowska, W.H.; Xu, J.; Abeysundara, N.; Leung, A.; Go, N.E.; DeVorkin, L.; Weppler, S.A.; Gelmon, K.; et al. Autophagy inhibition augments the anticancer effects of epirubicin treatment in anthracycline-sensitive and -resistant triple-negative breast cancer. Clin. Cancer Res. 2014, 20, 3159–3173. [Google Scholar] [CrossRef] [PubMed]
  47. McAfee, Q.; Zhang, Z.; Samanta, A.; Levi, S.M.; Ma, X.H.; Piao, S.; Lynch, J.P.; Uehara, T.; Sepulveda, A.R.; Davis, L.E.; et al. Autophagy inhibitor Lys05 has single-agent antitumor activity and reproduces the phenotype of a genetic autophagy deficiency. Proc. Natl. Acad. Sci. USA 2012, 109, 8253–8258. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, J.W.; Zhang, S.S.; Song, J.R.; Sun, K.; Zong, C.; Zhao, Q.D.; Liu, W.T.; Li, R.; Wu, M.C.; Wei, L.X. Autophagy inhibition switches low-dose camptothecin-induced premature senescence to apoptosis in human colorectal cancer cells. Biochem. Pharmacol. 2014, 90, 265–275. [Google Scholar] [CrossRef] [PubMed]
  49. Yuan, H.; Li, A.J.; Ma, S.L.; Cui, L.J.; Wu, B.; Yin, L.; Wu, M.C. Inhibition of autophagy signi fi cantly enhances combination therapy with sorafenib and HDAC inhibitors for human hepatoma cells. World J. Gastroenterol. 2014, 20, 4953–4962. [Google Scholar] [CrossRef] [PubMed]
  50. Sun, Y.; Liu, J.H.; Jin, L.; Sui, Y.X.; Lai, L.; Yang, Y. Inhibition of beclin 1 expression enhances cisplatin-induced apoptosis through a mitochondrial-dependent pathway in human ovarian cancer SKOV3/DDP cells. Oncol. Res. 2014, 21, 261–269. [Google Scholar] [CrossRef] [PubMed]
  51. Liang, X.; Tang, J.; Liang, Y.; Jin, R.; Cai, X. Suppression of autophagy by chloroquine sensitizes 5-fluorouracil-mediated cell death in gallbladder carcinoma cells. Cell Biosci. 2014, 4, 10. [Google Scholar] [CrossRef] [PubMed]
  52. Ji, C.; Zhang, L.; Cheng, Y.; Patel, R.; Wu, H.; Zhang, Y.; Wang, M.; Ji, S.; Belani, C.P.; Yang, J.M.; Ren, X. Induction of autophagy contributes to crizotinib resistance in ALK-positive lung cancer. Cancer Biol. Ther. 2014, 15, 570–577. [Google Scholar] [CrossRef] [PubMed]
  53. Yang, C.; Gao, R.; Wang, J.; Yuan, W.; Wang, C.; Zhou, X. High-mobility group nucleosome-binding domain 5 increases drug resistance in osteosarcoma through upregulating autophagy. Tumour Biol. 2014, 35, 6357–6363. [Google Scholar] [CrossRef] [PubMed]
  54. Nguyen, H.G.; Yang, J.C.; Kung, H.J.; Shi, X.B.; Tilki, D.; Lara, P.N., Jr.; Devere White, R.W.; Gao, A.C.; Evans, C.P. Targeting autophagy overcomes Enzalutamide resistance in castration-resistant prostate cancer cells and improves therapeutic response in a xenograft model. Oncogene 2014, 33, 4521–4530. [Google Scholar] [CrossRef] [PubMed]
  55. Gong, J.; Munoz, A.R.; Chan, D.; Ghosh, R.; Kumar, A.P. STAT3 down regulates LC3 to inhibit autophagy and pancreatic cancer cell growth. Oncotarget 2014, 5, 2529–2541. [Google Scholar] [PubMed]
  56. Zhai, B.; Hu, F.; Jiang, X.; Xu, J.; Zhao, D.; Liu, B.; Pan, S.; Dong, X.; Tan, G.; Wei, Z.; et al. Inhibition of Akt reverses the acquired resistance to sorafenib by switching protective autophagy to autophagic cell death in hepatocellular carcinoma. Mol. Cancer Ther. 2014, 13, 1589–1598. [Google Scholar] [CrossRef] [PubMed]
  57. Lu, Z.; Yang, H.; Sutton, M.N.; Yang, M.; Clarke, C.H.; Liao, W.S.; Bast, R.C., Jr. ARHI (DIRAS3) induces autophagy in ovarian cancer cells by downregulating the epidermal growth factor receptor, inhibiting PI3K and Ras/MAP signaling and activating the FOXo3a-mediated induction of Rab7. Cell Death Differ. 2014, 21, 1275–1289. [Google Scholar] [CrossRef] [PubMed]
  58. Lu, Z.; Baquero, M.T.; Yang, H.; Yang, M.; Reger, A.S.; Kim, C.; Levine, D.A.; Clarke, C.H.; Liao, W.S.; Bast, R.C., Jr. DIRAS3 regulates the autophagosome initiation complex in dormant ovarian cancer cells. Autophagy 2014, 10, 1071–1092. [Google Scholar] [CrossRef] [PubMed]
  59. Monti, M.C.; Margarucci, L.; Riccio, R.; Bonfili, L.; Mozzicafreddo, M.; Eleuteri, A.M.; Casapullo, A. Mechanistic insights on petrosaspongiolide M inhibitory effects on immunoproteasome and autophagy. Biochim. Biophys. Acta 2014, 1844, 713–721. [Google Scholar] [CrossRef] [PubMed]
  60. Yadav, A.K.; Vashishta, V.; Joshi, N.; Taneja, P. AR-A 014418 used against GSK3beta downregulates expression of hnRNPA1 and SF2/ASF splicing factors. J. Oncol. 2014, 2014, 695325. [Google Scholar] [CrossRef] [PubMed]
  61. Carr, G.; Williams, D.E.; Diaz-Marrero, A.R.; Patrick, B.O.; Bottriell, H.; Balgi, A.D.; Donohue, E.; Roberge, M.; Andersen, R.J. Bafilomycins produced in culture by Streptomyces spp. isolated from marine habitats are potent inhibitors of autophagy. J. Nat. Prod. 2010, 73, 422–427. [Google Scholar] [CrossRef] [PubMed]
  62. Kallifatidis, G.; Hoepfner, D.; Jaeg, T.; Guzman, E.A.; Wright, A.E. The marine natural product manzamine A targets vacuolar ATPases and inhibits autophagy in pancreatic cancer cells. Mar. Drugs 2013, 11, 3500–3516. [Google Scholar] [CrossRef] [PubMed]
  63. Guzman, E.A.; Johnson, J.D.; Linley, P.A.; Gunasekera, S.E.; Wright, A.E. A novel activity from an old compound: Manzamine A reduces the metastatic potential of AsPC-1 pancreatic cancer cells and sensitizes them to TRAIL-induced apoptosis. Invest. New Drugs 2011, 29, 777–785. [Google Scholar] [CrossRef] [PubMed]
  64. Kanno, S.; Yomogida, S.; Tomizawa, A.; Yamazaki, H.; Ukai, K.; Mangindaan, R.E.; Namikoshi, M.; Ishikawa, M. Papuamine causes autophagy following the reduction of cell survival through mitochondrial damage and JNK activation in MCF-7 human breast cancer cells. Int. J. Oncol. 2013, 43, 1413–1419. [Google Scholar] [PubMed]
  65. Yamazaki, H.; Wewengkang, D.S.; Kanno, S.; Ishikawa, M.; Rotinsulu, H.; Mangindaan, R.E.; Namikoshi, M. Papuamine and haliclonadiamine, obtained from an Indonesian sponge Haliclona sp., inhibited cell proliferation of human cancer cell lines. Nat. Prod. Res. 2013, 27, 1012–1015. [Google Scholar] [CrossRef] [PubMed]
  66. Keyzers, R.A.; Daoust, J.; Davies-Coleman, M.T.; van Soest, R.; Balgi, A.; Donohue, E.; Roberge, M.; Andersen, R.J. Autophagy-modulating aminosteroids isolated from the sponge Cliona celata. Org. Lett. 2008, 10, 2959–2962. [Google Scholar] [CrossRef] [PubMed]
  67. Liu, W.K.; Ling, Y.H.; Cheung, F.W.; Che, C.T. Stellettin A induces endoplasmic reticulum stress in murine B16 melanoma cells. J. Nat. Prod. 2012, 75, 586–590. [Google Scholar] [CrossRef] [PubMed]
  68. Liu, W.K.; Cheung, F.W.; Che, C.T. Stellettin A induces oxidative stress and apoptosis in HL-60 human leukemia and LNCaP prostate cancer cell lines. J. Nat. Prod. 2006, 69, 934–937. [Google Scholar] [CrossRef] [PubMed]
  69. Li, D.D.; Guo, J.F.; Huang, J.J.; Wang, L.L.; Deng, R.; Liu, J.N.; Feng, G.K.; Xiao, D.J.; Deng, S.Z.; Zhang, X.S.; Zhu, X.F. Rhabdastrellic acid-A induced autophagy-associated cell death through blocking Akt pathway in human cancer cells. PLoS One 2010, 5, e12176. [Google Scholar] [CrossRef] [PubMed]
  70. Guo, J.F.; Zhou, J.M.; Zhang, Y.; Deng, R.; Liu, J.N.; Feng, G.K.; Liu, Z.C.; Xiao, D.J.; Deng, S.Z.; Zhu, X.F. Rhabdastrellic acid-A inhibited PI3K/Akt pathway and induced apoptosis in human leukemia HL-60 cells. Cell Biol. Int. 2008, 32, 48–54. [Google Scholar] [CrossRef] [PubMed]
  71. Paul, S.; Kundu, R. Antiproliferative activity of methanolic extracts from two green algae, Enteromorpha intestinalis and Rhizoclonium riparium on HeLa cells. Disab. Advocacy Resour. Unit 2013, 21, 72. [Google Scholar]
  72. Dos Santos, A.O.; Veiga-Santos, P.; Ueda-Nakamura, T.; Filho, B.P.; Sudatti, D.B.; Bianco, E.M.; Pereira, R.C.; Nakamura, C.V. Effect of elatol, isolated from red seaweed Laurencia dendroidea, on Leishmania amazonensis. Mar. Drugs 2010, 8, 2733–2743. [Google Scholar]
  73. Campos, A.; Souza, C.B.; Lhullier, C.; Falkenberg, M.; Schenkel, E.P.; Ribeiro-do-Valle, R.M.; Siqueira, J.M. Anti-tumour effects of elatol, a marine derivative compound obtained from red algae Laurencia microcladia. J. Pharm. Pharmacol. 2012, 64, 1146–1154. [Google Scholar] [CrossRef] [PubMed]
  74. Hou, L.L.; Gao, C.; Chen, L.; Hu, G.Q.; Xie, S.Q. Essential role of autophagy in fucoxanthin-induced cytotoxicity to human epithelial cervical cancer HeLa cells. Acta Pharmacol. Sin. 2013, 34, 1403–1410. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, J.; Chen, S.; Xu, S.; Yu, X.; Ma, D.; Hu, X.; Cao, X. In vivo induction of apoptosis by fucoxanthin, a marine carotenoid, associated with down-regulating STAT3/EGFR signaling in sarcoma 180 (S180) xenografts-bearing mice. Mar. Drugs 2012, 10, 2055–2068. [Google Scholar] [CrossRef] [PubMed]
  76. Zhu, K.; Dunner, K., Jr.; McConkey, D.J. Proteasome inhibitors activate autophagy as a cytoprotective response in human prostate cancer cells. Oncogene 2010, 29, 451–462. [Google Scholar] [CrossRef] [PubMed]
  77. Ahn, K.S.; Sethi, G.; Chao, T.H.; Neuteboom, S.T.; Chaturvedi, M.M.; Palladino, M.A.; Younes, A.; Aggarwal, B.B. Salinosporamide A (NPI-0052) potentiates apoptosis, suppresses osteoclastogenesis, and inhibits invasion through down-modulation of NF-kappaB regulated gene products. Blood 2007, 110, 2286–2295. [Google Scholar] [CrossRef] [PubMed]
  78. Yang, F.; Chen, W.D.; Deng, R.; Li, D.D.; Wu, K.W.; Feng, G.K.; Li, H.J.; Zhu, X.F. Hirsutanol A induces apoptosis and autophagy via reactive oxygen species accumulation in breast cancer MCF-7 cells. J. Pharmacol. Sci. 2012, 119, 214–220. [Google Scholar] [CrossRef] [PubMed]
  79. Yang, F.; Chen, W.D.; Deng, R.; Zhang, H.; Tang, J.; Wu, K.W.; Li, D.D.; Feng, G.K.; Lan, W.J.; Li, H.J.; et al. Hirsutanol A, a novel sesquiterpene compound from fungus Chondrostereum sp., induces apoptosis and inhibits tumor growth through mitochondrial-independent ROS production: Hirsutanol A inhibits tumor growth through ROS production. J. Transl. Med. 2013, 11, 32. [Google Scholar] [CrossRef] [PubMed]
  80. Zhao, Y.; Chen, H.; Shang, Z.; Jiao, B.; Yuan, B.; Sun, W.; Wang, B.; Miao, M.; Huang, C. SD118-xanthocillin X (1), a novel marine agent extracted from Penicillium commune, induces autophagy through the inhibition of the MEK/ERK pathway. Mar. Drugs 2012, 10, 1345–1359. [Google Scholar] [CrossRef] [PubMed]
  81. Hau, A.M.; Greenwood, J.A.; Lohr, C.V.; Serrill, J.D.; Proteau, P.J.; Ganley, I.G.; McPhail, K.L.; Ishmael, J.E. Coibamide A induces mTOR-independent autophagy and cell death in human glioblastoma cells. PLoS One 2013, 8, e65250. [Google Scholar] [CrossRef] [PubMed]
  82. Randazzo, A.; Debitus, C.; Minale, L.; Garcia Pastor, P.; Alcaraz, M.J.; Paya, M.; Gomez-Paloma, L. Petrosaspongiolides M–R: New potent and selective phospholipase A2 inhibitors from the New Caledonian marine sponge Petrosaspongia nigra. J. Nat. Prod. 1998, 61, 571–575. [Google Scholar] [CrossRef]
  83. Forestieri, R.; Donohue, E.; Balgi, A.; Roberge, M.; Andersen, R.J. Synthesis of clionamine B, an autophagy stimulating aminosteroid isolated from the sponge Cliona celata. Org. Lett. 2013, 15, 3918–3921. [Google Scholar] [CrossRef] [PubMed]
  84. Tang, B.; Cai, J.; Sun, L.; Li, Y.; Qu, J.; Snider, B.J.; Wu, S. Proteasome inhibitors activate autophagy involving inhibition of PI3K-Akt-mTOR pathway as an anti-oxidation defense in human RPE cells. PLoS One 2014, 9, e103364. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, G.Y.S.; Abrell, L.M.; Avelar, A.; Borgeson, B.M.; Crews, P. New hirsutane based sesquiquiterpenes from salt water cultures of a marine sponge-derived fungus and the terrestrial fungus coriolus consors. Tetrahedron 1998, 54, 7335–7342. [Google Scholar] [CrossRef]
  86. Yao, Q.H.; Zhang, X.C.; Fu, T.; Gu, J.Z.; Wang, L.; Wang, Y.; Lai, Y.B.; Wang, Y.Q.; Guo, Y. omega-3 polyunsaturated fatty acids inhibit the proliferation of the lung adenocarcinoma cell line A549 in vitro. Mol. Med. Rep. 2014, 9, 401–406. [Google Scholar] [PubMed]
  87. Ivanchina, N.V.; Kicha, A.A.; Stonik, V.A. Steroid glycosides from marine organisms. Steroids 2011, 76, 425–454. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, Y.; Zhan, Y.; Xu, R.; Shao, R.; Jiang, J.; Wang, Z. Src mediates extracellular signal-regulated kinase 1/2 activation and autophagic cell death induced by cardiac glycosides in human non-small cell lung cancer cell lines. Mol. Carcinog. 2014, in press [PMID: 24610665]. [Google Scholar]

Share and Cite

MDPI and ACS Style

Farooqi, A.A.; Fayyaz, S.; Hou, M.-F.; Li, K.-T.; Tang, J.-Y.; Chang, H.-W. Reactive Oxygen Species and Autophagy Modulation in Non-Marine Drugs and Marine Drugs. Mar. Drugs 2014, 12, 5408-5424. https://doi.org/10.3390/md12115408

AMA Style

Farooqi AA, Fayyaz S, Hou M-F, Li K-T, Tang J-Y, Chang H-W. Reactive Oxygen Species and Autophagy Modulation in Non-Marine Drugs and Marine Drugs. Marine Drugs. 2014; 12(11):5408-5424. https://doi.org/10.3390/md12115408

Chicago/Turabian Style

Farooqi, Ammad Ahmad, Sundas Fayyaz, Ming-Feng Hou, Kun-Tzu Li, Jen-Yang Tang, and Hsueh-Wei Chang. 2014. "Reactive Oxygen Species and Autophagy Modulation in Non-Marine Drugs and Marine Drugs" Marine Drugs 12, no. 11: 5408-5424. https://doi.org/10.3390/md12115408

Article Metrics

Back to TopTop