You are on page 1of 9

Received: 13 January 2019 | Revised: 3 April 2019 | Accepted: 11 April 2019

DOI: 10.1002/jcp.28740

REVIEW ARTICLE

Autophagy, anoikis, ferroptosis, necroptosis, and endoplasmic


reticulum stress: Potential applications in melanoma therapy

Milad Ashrafizadeh1 | Reza Mohammadinejad2 | Shima Tavakol3 |


Zahra Ahmadi4 | Sahar Roomiani4 | Majid Katebi5

1
Department of Basic Science, Faculty of
Veterinary Medicine, University of Tabriz, Abstract
Tabriz, Iran Melanoma as the most major skin malignancy has attracted much attention, so far.
2
Pharmaceutics Research Center, Institute of
Although a successful therapeutic strategy requires an accurate understanding of the
Neuropharmacology, Kerman University of
Medical Sciences, Kerman, Iran precise mechanisms for the initiation and progression of the melanoma. Several types
3
Cellular and Molecular Research Center, Iran of cell death mechanisms have recently been identified along with conventional cell
University of Medical Sciences, Tehran, Iran
4
death mechanisms such as apoptosis and necrosis. Among those mechanisms,
Department of Basic Science, Faculty of
Veterinary Medicine, Islamic Azad Branch, necroptosis, anoikis, ferroptosis, and autophagy may be considered to have
University of Shushtar, Khuzestan, Iran
remarkable modulatory impacts on melanoma. In the present review, we explain
5
Department of Anatomy, Hormozgan
University of Medical Sciences, Bandar Abbas,
the mechanisms of cell death signaling pathways related to autophagy, ferroptosis,
Hormozgan, Iran anoikis, necroptosis, and reticulum endoplasmic stress in cells and describe how those

Correspondence
mechanisms transduce signals in melanoma cells. Meanwhile, we describe how we can
Milad Ashrafizadeh, Department of basic modulate those mechanisms to eliminate melanoma.
science, Faculty of Veterinary Medicine,
University of Tabriz, Tabriz, Iran.
KEYWORDS
Email: dvm.milad73@yahoo.com
anoikis, autophagy, ferroptosis, melanoma., necroptosis

1 | INTRODUCTION important role of regulated cell death (RCD) in preserving homeostasis


during physiological and pathological conditions (Galluzzi, López‐Soto,
Melanoma, the most common cause of skin malignancy, is resulted from Kumar, & Kroemer, 2016). Briefly, cell death is divided into three major
malignant transformation of melanocytes. It is thought that the categories including (1) type I programmed cell death or apoptosis; (2)
increase of melanoma incidence particularly in western countries is in type II programmed cell death or autophagy; and (3) type III accidental
part owing to their brighter skin and lower sun protection (Gallagher, cell death or necrosis (Shakeri, Cicero, Panahi, Mohajeri, & Sahebkar,
2005). Besides, lack of a sensitive and precise approaches to diagnose 2018). To effectively remedy melanoma, treatment strategies should be
melanoma at the early stages leads to cancer progression and not focused on other cell death mechanisms instead of apoptosis. This is
properly responding to the therapeutics (Aris & Barrio, 2015; Tarver, due to the proliferative effect of apoptosis on neighboring surviving
2012). Although there are some promising investigations on the cells, a phenomenon known as apoptosis‐induced proliferation (AiP). In
efficacy of novel nano‐therapeutics on skin repair (Tavakol, Jalili‐ recent years, it has been shown that apoptosis can induce in a paracrine
Firoozinezhad, Mashinchian, & Mahmoudi, 2016; Tavakol, Zare, manner a proliferative response in neighboring surviving cells. This
Hoveizi, Tavakol, & Rezayat, 2019), during recent years, much attention phenomenon has considerable clinical implications because apoptotic
has been made to kill melanoma cells without affecting healthy cells. By treatments can induce cell proliferation and cancer stem cell recruit-
regards to the importance of cancer therapy and tumor cell mortality, ment in different cancer types such as melanoma (Hurle, 2014). Donato
the precise understanding of the processes involved in cell death is et al. (2014) demonstrated the induction of caspase 3‐dependent cell
vital. Research works have manifested a series of pathways which are death in melanoma cells leads to the proliferation of melanoma cells. It
involved in the cell homeostasis to eliminate irreversible damaged cells was shown that caspase 3‐mediated dying melanoma cells stimulates
including cells with excessive and toxic biomolecules, malfunction the growth and proliferation of melanoma cells through the secretion
organelles, etc. (Galluzzi et al., 2018). Multiple studies showed the of prostaglandin E2 (PGE2). In another study conducted by Roumane,
J Cell Physiol. 2019;234:19471–19479. wileyonlinelibrary.com/journal/jcp © 2019 Wiley Periodicals, Inc. | 19471
19472 | ASHRAFIZADEH ET AL.

Berthenet, El Fassi, and Ichim (2018), the stimulatory effect of caspase‐ The role of autophagy in cancer is sophisticated and is dependent to
dependent cell death on the proliferation of melanoma cells was the tumor type and driving oncogene (Nyfeler & Eng, 2016).
confirmed. It was revealed that an alternative kind of cell death, known Previously, it was thought that autophagy has a remarkable tumor‐
as caspase‐independent cell death (CICD), has inhibitory effect on the suppressive role, whereas now it is believed that autophagy plays a
proliferation of melanoma cells and remarkably suppresses the significant protumorigenic role in a number of cancers. However, the
migration of tumor cells, whereas, apoptotic cell death is able to stage of cancer dictates the role of autophagy as an antitumor or
induce the proliferation of living melanoma cells, requiring a novel and protumorogenic mechanisms (Rabiee, Tavakol, Barati, & Joghataei,
revision of the therapeutic strategies based on other cell death 2018). Several studies exhibit that autophagy can be considered as a
pathways. survival mechanism in cancer cells against therapies such as
In the present review, we focus on the influence of cell death chemotherapy and radiotherapy (Rebecca & Amaravadi, 2016).
mechanisms involved in melanoma therapy and will discuss how to Eventually, it promotes durability of cancer cells against apoptosis
sensitize melanoma cells with chemotherapeutics. (Fitzwalter et al., 2018). Thorburn et al. (2014) demonstrated that
following autophagy inhibition, proapoptotic protein PUMA (activa-
tor of apoptosis) increases. Thus, modulation of autophagy can be
2 | AUT O PHA GY M A CH IN E considered as a promising strategy to diminish cancer progression.

Autophagy was coined in 1950s following the mammalian cells


observation using electron microscopy (Mohammadinejad, Ahmadi, 3 | AN OIKIS
Tavakol, & Ashrafizadeh, 2019). This mechanism is mainly divided into
three main categories including microautophagy, macroautophagy, and Anoikis was introduced for the first time in epithelial and endothelial cells
chaperone‐mediated autophagy (Kimmelman & White, 2017; Shakeri (Terasaki et al., 2018). It is a special type of apoptotic cell death resulting
et al., 2018). Briefly, in microautophagy, cytoplasmic cargos are from the lack of cell attachment to the extracellular matrix (ECM) or
engulfed in lysosomal or endosomal membranes, whereas chaperone‐ adhesion to wrong locations, therefore, it mainly depends on the cell–
mediated autophagy as a selective degradation process special to matrix interactions (Kakavandi, Shahbahrami, Goudarzi, Eslami, &
mammalian cells, targets proteins containing KFERQ‐like motif Faghihloo, 2018). Anoikis is critical for development and tissue home-
(Cuervo & Wong, 2014). The most well‐known type of autophagy is ostasis. It was demonstrated that this important pathway inhibits tumor
macroautophagy in which intracellular components are surrounded by formation by neoplastic cells (Gilmore, 2005); however, tumor cells are
the double‐membrane compartments, known as autophagosome, and resistant to anoikis through TrkB (Mawji et al., 2007). To sum things up,
then, their content releases into lysosome where the degradation and cell detachment from ECM leads to anoikis, cell proliferation in
recycling processes occur (Klionsky & Codogno, 2013). Noteworthy, inappropriate locations, and metastasis in long‐range organs (Chiarugi &
despite the great amount of investigations in neurological diseases Giannoni, 2008; Frisch & Screaton, 2001). A number of intrinsic and
(Abdolmaleky et al., 2015; Faghihi et al., 2016; Tavakol et al., 2017; extrinsic signaling pathways are engaged with the activation of anoikis
Zendedel et al., 2012), impairment or decline in autophagy has been and its progression (Grossmann, 2002). The intrinsic pathway mainly
observed in ageing and neurodegenerative diseases such as amyo- crosses through the mitochondria. Briefly, following stress conditions
trophic lateral sclerosis (Menzies, Fleming, & Rubinsztein, 2015). such as DNA damage and ER stress, Bax, and Bak are transmitted from
Autophagy has dual effects on cells, it acts as both cell survival and the cytosol to outer mitochondria membrane (OMM) and increases
lethal mechanisms to modulate homeostasis in cells (White, 2016). In mitochondrial permeabilization and cytochrome c release. Then, cyto-
other words, autophagy is activated under stress conditions such as chrome c forms apoptosome and as a consequence, apoptosis occurs
starvation and acidic pH microenvironment to guarantee cell survival (Cohen, 1997). BH3‐only proteins are considered as the crucial mediators
(Tavakol, 2014), because in physiological conditions, it acts at the during anoikis. Upon the cell‐ECM detachment, Bid, and Bam (members
baseline level to prevent the accumulation of excessive biomolecules of BH3‐only proteins) are activated and enhances the oligomerization of
and damaged cells and organelles. Autophagy performs through a Bax‐Bak inside the OMM (Taylor, Cullen, & Martin, 2008). However, Bad,
variety of proteins encoded by autophagy‐related genes (ATGs). It has Bik, Bmf, Noxa, Puma, and Hrk are the other members of BH3‐only family
been reported that more than 30 ATGs are involved in autophagy and indirectly are involved in the oligomerization of Bax‐Bak (Kuwana
(Klionsky et al., 2011). Upon the insufficient level of energy (ATP et al., 2005). On the other hand, various death receptor transmembrane
form), AMP‐activated protein kinase (AMPK) is activated that proteins such as Fas receptor, TNFR1, and TNF‐related apoptosis
consequently leads to autophagy induction. However, mammalian inducing ligand (TRAIL) receptor‐1 and ‐2 are engaged with the death
target of rapamycin (mTOR) negatively is involved in autophagy signaling pathways. Following the ligand binding, death‐inducing signaling
regulation, as well. Activation of mTORC1 inhibits autophagy and complex (DISC) is formed and it increases the activation of a number of
under the lack of growth factors and/or amino acids, autophagy is molecules including caspase‐8 through interplay with Fas‐associated
triggered due to the inhibition of mTORC1. death domain protein (FADD). Finally, activation of caspase‐8 leads to
Nowadays, autophagy has attracted much attention owing to its induction of caspase‐3, ‐6, and ‐7 and subsequently, cell death
major role in cancer progression and therapy (Galluzzi et al., 2015). (Mohammadinejad et al., 2018). It has been shown that upon the cel
ASHRAFIZADEH ET AL. | 19473

ECM detachment, Fas and Fas ligand are upregulated, demonstrating the death is different from apoptosis, classic necrosis, autophagy, and
vital role of extrinsic pathway in anoikis execution (Aoudjit & Vuori, other types of cell death (Dixon et al., 2012). Intracellular ROS
2001). production and lipid peroxides accumulation in cells are the
Anoikis regulation is a crucial step in cancer therapy owing to its hallmarks of ferroptosis (Xie et al., 2016). In other words, ferroptosis
critical role in cancer metastasis. In a study, Chen et al. (2018) is activated through the high levels of iron in a process which known
examined the effect of hepatocellular carcinoma‐related protein‐1 as Fenton reaction (Yang et al., 2014) and through the ROS
(HCRP‐1) on human colon cancer. Their results showed that HCRP‐1 accumulation resulting from impairment in cellular glutathione‐
inhibition is associated with tumor metastasis and it was found that dependent antioxidant defenses (Mawji et al. 2007). To sum things
HCRP‐1 positively affects cell anoikis through Bim regulation. up, ROS production, nicotinamide adenine dinucleotide phosphate
Notably, the regulation of anoikis via HCRP‐1 can be considered as (NADPH)‐dependent lipid peroxidation and GSH depletion stimulate
a suitable strategy in colon cancer therapy. Taking everything into ferroptosis (Yang et al., 2014). Furthermore, the family of mitogen‐
account, these findings disclosed the major role of anoikis in tumor activated protein kinase (MAPK) including ERK, P38, and c‐Jun NH2‐
metastasis and survival and shed light in cancer biology and therapy. terminal kinase (JNK) are involved in ferroptosis. The morphological
characteristics of ferroptosis are including intact cell membrane,
ordinary volume of nucleus and dense miniature mitochondria which
4 | NEC ROP TOSIS are distinct from necrosis and apoptosis (Dolma, Lessnick, Hahn, &
Stockwell, 2003). It has been demonstrated that ferroptosis is
Necroptosis was introduced in 2005 by Dagterev et al. (Li, Ning, Lou, & engaged with a great number of pathological conditions such as cell
Xu, 2018) as a new form of programmed cell death with similar death in kidney, brain and heart I/R injuries with the high levels of
morphological characteristic to necrosis (Nikseresht, Khodagholi, & glutamate (Angeli et al., 2014). However, the role of ferroptosis in
Ahmadiani, 2018). It has some features of apoptosis and necrosis; for cancer is crucial and there are studies indicated in the tumor‐
example, it has morphological signs such as early disruption of plasma suppressive role of ferroptosis (Yang et al., 2014). Taking everything
membrane, translucent cytosol, enhanced cell size, and swollen into account, the understanding of the ferroptosis mechanisms and
organelles (Ofengeim & Yuan, 2013) whereas it can be reversed like interactions in cancer is of interest in terms of developing novel
apoptosis. Necroptosis is related to the various pathological conditions diagnostic and therapeutic approaches to adjust cell survival and
such as ischemia/reperfusion (I/R) injury, nonalcoholic steatohepatitis, death in pathological conditions.
sepsis, and atherosclerosis (Shan, Pan, Najafov, & Yuan, 2018). Although
it has a remarkable role in the efficacy of a number of cancer
therapeutics (Cruz, Qin, Stewart, & Chen, 2018), the belief is that 6 | ENDOPLASMIC RETICULUM STRESS
multiple signaling pathways are involved in activation of necroptosis
which are induced via numerous stimuli (Mohammadinejad et al., 2018). Endoplasmic reticulum (ER) as a largest organelle in eukaryotic cells, is a
Necroptosis is a caspase‐independent mechanism involving in the double‐membrane compartment and is engaged with protein synthesis,
activation of death receptors (Galluzzi, Kepp, Chan, & Kroemer, 2017). folding and maturation of approximately one‐third of the eukaryotic
During necroptosis, receptor interacting serine/threonine kinase 3 proteins (Mollazadeh et al., 2018). However, ER is an organelle for Ca2+
(PIRK3) and substrate mixed lineage kinase domain‐like (MLKL) play storage and release, and lipid synthesis (Villalobos‐Labra, Subiabre,
significant roles in activation and execution of cell death, respectively Toledo, Pardo, & Sobrevia, 2018); therefore, ER homeostasis is vital for
(Hadifar et al., 2018). Following the phosphorylation of MLKL by PIRK3, cells. The imbalance between folding capacity of ER and newly
MLKL is oligomerized and translocated to the plasma membrane, where synthesized proteins leads to ER stress (Gallazzini & Pallet, 2018). Unfold
it enhances the permeability of membrane through the interaction with protein response (UPR) is one of the critical mechanisms involved in the
a variety of phospholipids. This permeabilization is the major difference quality monitoring of ER. UPR promotes ER homeostasis through three
between apoptosis and necrosis. To further clarify the necroptosis major signaling pathways which are integrated in the ER membrane:
phenomena and its difference with apoptosis and necrosis, it may be Inositol‐requiring protein1 (IRE1), activating transcription factor 6 (ATF6),
said that apoptotic cells are surrounded by antigen‐presenting cells or and PRK‐like ER kinase (PERK; Walter & Ron, 2011). IRE1 interferes with
by adjacent cells, whereas in necroptosis, permeabilization prompts RNA (enhances RNA degradation) to diminish protein synthesis (Hollien
releases of cytokines, and chemokines, resulting in the induction of & Weissman, 2006). IRE1 indirectly participates in UPR and upon its
inflammation and immune response (Pasparakis & Vandenabeele, 2015). activation, it produces spliced form of X box‐binding protein 1 (sXBP1)
and then, sXBP1 activates genes that contribute to alleviating ER stress
to restore ER folding capacity (Acosta‐Alvear et al., 2007). On the other
5 | FERROPTOSIS hand, PERK is associated with transcription factors namely nuclear factor
erythroid 2‐related factor 2 (NRF2) and eukaryotic translation initiation
Another type of programmed cell death is ferroptosis with the factor 2 (elF2α). NRF2 stimulates antioxidant proteins to regulate
involvement of iron which was introduced by Dr. Brent R. Stockwell oxidative stress whereas elF2α is involved in mRNA translation. Upon
in 2012 (Latunde‐Dada, 2017). This oxidative and iron‐mediated cell elF2α inhibition, protein synthesis is diminished. Furthermore, during ER
19474 | ASHRAFIZADEH ET AL.

F I G U R E 1 The induction of apoptosis, ER stress, anoikis, necroptosis, and autophagy using special drugs lead to melanoma inhibition. Noteworthy,
the inhibition and enhancement of autophagy have the two‐fold role in melanoma elimination. [Color figure can be viewed at wileyonlinelibrary.com]

stress, ATF6 translocates to Golgi apparatus to release transcription P38 MAPK‐selective inhibitor (SB202190) enhances apoptosis and
factor (Shen, Chen, Hendershot, & Prywes, 2002). AFT6 targets UPR suppresses ALS‐induced autophagy. Noteworthy, autophagy inhibition
proteins such as chaperons and also stimulates XBP1 and C/EBP is associated with the high sensitization of melanoma cells to ALS‐
homologous protein (CHOP) genes (Yoshida et al., 2000). induced apoptosis. It has been reported that autophagy‐defective
It has been shown that ER stress stimulates autophagy and tumors are not capable to recruit natural killer (NK) cells. Therefore,
apoptosis. ER‐associated degradation (ERAD) and UPR are the autophagy targeting in melanoma cells may diminish tumor growth via
quality control mechanisms of ER which contribute to ubiquition recruiting NK cells infiltration (Mgrditchian et al., 2017).
and degradation of misfolded proteins via proteasome and autophagy Meanwhile, Luan et al. (2017) investigated the effect of
(Gu et al., 2017). At the mild and acute ER stress, autophagy is a polygonatum odoratum lectin (POL) on autophagy in melanoma cells.
dominate process compared with the apoptosis whereas following Their results demonstrated that POL decreases melanoma cell
prolonged and exacerbate ER stress, apoptosis is triggered and cells survival through downregulation of miR1290, leading to BECN1
decide to suicide (B’chir et al., 2013). ER stress triggers autophagy upregulation and autophagy enhancement. Xiao, Diao, Liang, Peng,
through the AMPK activation (Meares et al., 2011). As be mentioned and Zeng (2017) studied the effect of miR‐24‐1‐5p as an autophagy
earlier, three major transmembrane proteins are involved in UPR inducer in melanoma cells. They found that miR‐24‐1‐5p remarkably
including IRE1, ATF6, and PERK. Furthermore, under ER stress, ATF6 enhances the levels of LC3‐II/I ratio and beclin‐1 expression,
positively affects the expression of death‐associated protein kinase 1 consequently resulting in autophagy promotion and the inhibition
(DAPK1), consequently resulting in autophagy induction (Zalckvar of melanoma proliferation. Moreover, proopiomelanocortin (POMC)
et al., 2009). Noteworthy, CHOP is involved in PERK and IRE1 induces melanoma cell elimination through the autophagy‐mediated
pathways to induce apoptosis (Bu & Diehl, 2016). It has been cell death (Wu, Tsai, Liu, Wu, & Tai, 2018; Figure 1).
reported that protease, kinase, transcription factors, Bcl‐2 family
proteins as well as their modulators are involved in the triggering of
apoptosis via ER stress‐mediated UPR (Mollazadeh et al., 2018). 8 | MELANO MA AND A NOIKIS

Anoikis resistant is a smart strategy in cancer cells to promote


7 | MELA NOMA AND AU TOPH AGY metastasis (P. Gupta, N. Gupta, Fofaria, Ranjan, & Srivastava, 2019).
It is worth nothing that anoikis‐resistant melanoma cells are not
Nowadays, much investigations have been made to understand the highly sensitive to chemotherapy (Tseng, Uen, Tseng, & Lee, 2017).
role of autophagy in cancer (Ahmadi, Mohammadinejad, & Ashrafiza- Hasnat, Pervin, Lim, and Lim (2015) investigated the effect of
deh, 2019; Mohammadinejad et al., 2019; Rabiee et al., 2018) and its apigenin on melanoma cells. They showed that apigenin decreases
relation to other cell death mechanisms such as ferroptosis (Gao et al., the level of integrin proteins (inhibitors of anoikis) and suppresses
2016). In a study, alisertib (ALS) was used to treat melanoma (Shang, the phosphorylation of focal adhesion kinase (FAK) and extracellular
Yao, & Zhou, 2017). It was demonstrated that ALS in combination with signal‐related kinase (ERK1/2), resulting in stimulation of anoikis in
ASHRAFIZADEH ET AL. | 19475

human melanoma cells (A2058 and A375 cell lines). Interestingly, it affect necroptosis in melanoma cells and also the inactivation of miR‐
has been shown that STAT3 enhances the invasiveness of melanoma 137 enhances the antitumor activity of erastin by elevating ferroptosis.
cells through anoikis inhibition (Fofaria & Srivastava, 2014). Beside of Moreover, it was demonstrated that mitochondrial complex I inhibitor is
STAT3, it has been reported that the upregulation of Timp1 makes an important target in the induction of ferroptosis in melanoma cells
cells resistant to anoikis. In an effort in line with this statement, the (Basit et al., 2017; Figure 1).
impact of Timp1 on melanoma cells was investigated (Toricelli, Melo,
Peres, Silva, & Jasiulionis, 2013). It was shown that Timp1 induces
anoikis resistance in melanoma cells through activating PI3‐k 11 | M E L ANO M A AN D EN D O PL AS M I C
signaling pathway. Furthermore, Goundiam, Nagel, and Vayssade RE T I C U LU M S T R ES S
(2012) investigated the effect of Akt and RhoA inhibition on the
metastasis of highly metastatic B16F10 melanoma cells. They found ER stress may be considered as a promising strategy to eliminate
that the inhibition of Akt and RhoA decreases the expression of α5 melanoma cells. There are some pharmaceuticals to diminish
integrin and inactivates focal adhesion kinase (FAK) in melanoma melanoma through the activation of ER stress‐induced apoptosis.
cells, leading to the anoikis induction in anoikis‐resistant cells. Other Honokiol‐mediated ER stress has been recognized to suppress the
studies were in agreement with the effect of anoikis on metastasis growth and metastasis of melanoma (Chiu et al., 2019). It induces
inhibition of melanoma cells (Boisvert‐Adamo & Aplin, 2006; apoptosis and inhibits epithelial‐mesenchymal transition (EMT) in
Boisvert‐Adamo, Longmate, Abel, & Aplin, 2009; Goldstein et al., highly metastatic melanoma xenograft mouse model via ER stress.
2009; Zhang et al., 2011; Zhu et al., 2001; Figure 1). Also, Heo, Kim, Hwang, Kang, and Choi (2018) investigated the effect
of resveratrol on ER stress‐induced apoptosis in A375SM malignant
melanoma cell line. They demonstrated that resveratrol suppresses
9 | MELA NOMA AND NEC ROP TOSIS the growth of melanoma cells via the stimulation of ER stress through
the overexpression of elF2‐α and CHOP. Furthermore, the antitumor
As the necroptosis is a newly recognized pathway in cancer cells, effect of pinocembrin on melanoma cells is based on ER stress (Zheng
therefore, there are a few studies to in detail explain its role and et al., 2018). It was exhibited that pinocembrin acts in a dose‐
effect in melanoma. Kong, Lv, Yan, Chang, and Wang (2018) dependent manner through IRE1a/Xbp1 pathway. Noteworthy, not
examined the effect of a novel naphthyridine derivative, 3u, on only ER stress induces apoptosis in melanoma cells but also, it sensitize
human melanoma A375 cells. It was shown that 3u stimulates melanoma cells to chemotherapy (Ryabaya et al., 2018). It has been
necroptosis at low concentrations through the upregulation of death demonstrated that the downregulation of ER stress chaperon 78 kDa
receptors and activation of caspase‐8. Furthermore, Basit et al. glucose‐regulated protein 78 (GRP78) prevents autophagy and
(2017) exhibited the necroptosis induction in melanoma cells through increases the effect of chemotherapy on melanoma cells. It has been
mitochondrial complex I inhibition. It has been shown that shown that bornyl cis‐4‐hydroxy cinnamate stimulates PERK and
necroptosis resistant in melanoma cells is related to the absence of elF2α pathways to activate ER stress‐apoptosis in melanoma cells
RIPK3 (Geserick et al., 2015). However, Hammerová, Uldrijan, (Yang, Wu, Chang, Chiu, & Wu, 2018). Interestingly, Heo, Lee, Kim,
Táborská, Vaculová, and Slaninová (2012) investigated the effect of Hwang, and Choi (2018) conducted a comprehensive experiment on
sanguilutine (SL) on melanoma cell mortality through the autophagy the impacts of kaempferol, genistein, and 3,3'‐diinolylmethane on ER
and necroptosis. It was found that SL stimulates caspase‐independent stress‐induced apoptosis on A375SM melanoma cells. Their results
non‐apoptotic cell death (necroptosis) whereas a combination of SL showed that these compounds increase the expression of elF2α and
and an autophagy inhibitor decreases the viability of melanoma cells, CHOP to exert ER stress‐mediated apoptosis in melanoma cells.
demonstrating that SL can be considered as an ideal compound in the Notwithstanding, mechano growth factor E peptide eliminates invasive
induction of necroptosis in melanoma cells and is a beneficial melanoma cells via inducing ER stress through the upregulation of cell
mediator to investigate the interplay between autophagy and apoptosis‐related protein CHOP (He et al., 2018; Figure 1).
necroptosis (Figure 1).

12 | CO N CL US I O N AND RE M AR K S
10 | MELANOMA AND FERRO PTOSIS
Melanoma as the most major skin malignancy has attracted much
Similar to necroptosis, ferroptosis is a newly introduced phenomenon in attention, so far. To have a successful therapeutic strategy for melanoma
melanoma and studies are increasing to manifest its role in melanoma. treatment, it requires an accurate understanding of the precise
In a study, Zhang et al. (2018) investigated the role of ferroptosis mechanisms involved in the initiation and the progression of melanoma.
regulation by miR‐9 in melanoma. They showed that miR‐9 decreases Recently, various cell death mechanisms beside of other conventional cell
the erastin‐ and RSL3‐induced ferroptosis in melanoma cells, demon- death mechanisms such as apoptosis and necrosis have been identified
strating that knocking down of miR‐9 induces ferroptosis in melanoma for the treatment of melanoma. The most major drawbacks of apoptosis
cells. Furthermore, Luo et al. (2018) exhibited that miR‐137 negatively backs on the stimulatory effect of apoptosis‐dependent cell death
19476 | ASHRAFIZADEH ET AL.

mechanisms on the growth and proliferation of surviving melanoma cells ORCI D


and also the impairment effect of CICD mechanisms on these tumor cells.
Milad Ashrafizadeh http://orcid.org/0000-0001-6605-822X
Among those mechanisms, necroptosis, anoikis, ferroptosis, and autop-
Shima Tavakol http://orcid.org/0000-0002-8531-7650
hagy may be considered to have the remarkable modulator impact on
melanoma. Although at the first view it seems that the induction of cell
death mechanisms in melanoma cells overcome to its progression, but in R E F E R E N CE S
fact, the story is a bit complicated in tumors and resistant to cell death
Abdolmaleky, H. M., Pajouhanfar, S., Faghankhani, M., Joghataei, M. T.,
mechanisms such as apoptosis, and anoikis leads to more investigation Mostafavi, A., & Thiagalingam, S. (2015). Antipsychotic drugs
with new approaches. The important note is related to the different attenuate aberrant DNA methylation of DTNBP1 (dysbindin) promo-
effects of chemotherapeutics associated with the different phases of ter in saliva and post‐mortem brain of patients with schizophrenia and
psychotic bipolar disorder. American Journal of Medical Genetics, Part B:
cancer and its invasiveness. It is interesting from one side, autophagy
Neuropsychiatric Genetics, 168(8), 687–696.
inhibition causes apoptosis in melanoma cells, whereas from other side, Acosta‐Alvear, D., Zhou, Y., Blais, A., Tsikitis, M., Lents, N. H., Arias, C., &
autophagy‐ defective tumors suffer from decreased recruiting of NK cells Dynlacht, B. D. (2007). XBP1 controls diverse cell type‐and condition‐
infiltration. Moreover, there are some reports indicating the promising specific transcriptional regulatory networks. Molecular Cell, 27(1), 53–66.
outcomes in the elimination of tumor cells through the targeting of Ahmadi, Z., Mohammadinejad, R., & Ashrafizadeh, M. (2019). Drug
delivery systems for resveratrol, a non‐flavonoid polyphenol: Emer-
autophagy mechanism. Notably, the most antimelanoma studies were in
ging evidence in last decades. Journal of Drug Delivery Science and
agreement with the induction of autophagy through BECN1 upregulation, Technology, 51, 591–604.
increasing the levels of LC3‐II/I ratio. In fact, cancer cells are considerably Angeli, J. P. F., Schneider, M., Proneth, B., Tyurina, Y. Y., Tyurin, V. A.,
smart and although are disconnected from the ECM and neighbor cells Hammond, V. J., & Eggenhofer, E. (2014). Inactivation of the
ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nature
but they do not become home sick and are resistant to anoikis and less
Cell Biology, 16(12), 1180–1191.
response to the chemotherapeutics. Noteworthy, the inhibition of AKT Aoudjit, F., & Vuori, K. (2001). Matrix attachment regulates Fas‐induced
and RhoA may be considered a promising strategy to induce anoikis in apoptosis in endothelial cells: A role for c‐flip and implications for
anoikis‐resistant melanoma cells. Besides, the necroptosis resistant anoikis. The Journal of Cell Biology, 152(3), 633–644.
Aris, M., & Barrio, M. M. (2015). Combining immunotherapy with
melanoma is in part due to the absence of RIPK3 and is modulated
oncogene‐targeted therapy: A new road for melanoma treatment.
through some pharmaceuticals such as naphthyridine derivative. Frontiers in Immunology, 6, 46.
Furthermore, it seems that knocking down of miR‐9 and 137 is a B’chir, W., Maurin, A.‐C., Carraro, V., Averous, J., Jousse, C., Muranishi, Y.,
promising strategy to induce ferroptosis and melanoma cell death. Taking & Bruhat, A. (2013). The eIF2α/ATF4 pathway is essential for stress‐
induced autophagy gene expression. Nucleic Acids Research, 41(16),
everything into account, designing of a successful strategy to eliminate
7683–7699.
cancer, needs to consider all aspects of the problem and also requires the Basit, F., Van Oppen, L. M., Schöckel, L., Bossenbroek, H. M., Van Emst‐de
use of all cell death mechanisms. Vries, S. E., Hermeling, J. C., & Willems, P. H. (2017). Mitochondrial
complex I inhibition triggers a mitophagy‐dependent ROS increase
leading to necroptosis and ferroptosis in melanoma cells. Cell Death &
CO NFLICT OF I NTERE STS Disease, 8(3), e2716.
Boisvert‐Adamo, K., & Aplin, A. (2006). B‐RAF and PI‐3 kinase signaling
Authors declare that they have no conflict of interests. protect melanoma cells from anoikis. Oncogene, 25(35), 4848–4856.
Boisvert‐Adamo, K., Longmate, W., Abel, E. V., & Aplin, A. E. (2009). Mcl‐1
is required for melanoma cell resistance to anoikis. Molecular Cancer
Research, 7(4), 549–556.
FUNDING
Bu, Y., & Diehl, J. A. (2016). PERK integrates oncogenic signaling and cell
survival during cancer development. Journal of Cellular Physiology,
There was no finding source to support the manuscript.
231(10), 2088–2096.
Chen, F., Zhang, L., Wu, J., Huo, F., Ren, X., Zheng, J., & Pei, D. (2018).
HCRP‐1 regulates EGFR–AKT–BIM‐mediated anoikis resistance and
serves as a prognostic marker in human colon cancer. Cell Death &
A UT HO RS CO NT RIB UT ION S
Disease, 9(12), 1176.
Chiarugi, P., & Giannoni, E. (2008). Anoikis: A necessary death program
MA conducted the manuscript and wrote the introduction, “Melano- for anchorage‐dependent cells. Biochemical Pharmacology, 76(11),
ma and necroptosis”, “Melanoma and ferroptosis,” and conclusion 1352–1364.
section. RM wrote the “Autophagy” and “Induction of autophagy and Chiu, C.‐S., Tsai, C.‐H., Hsieh, M.‐S., Tsai, S.‐C., Jan, Y.‐J., Lin, W.‐Y., & Arbiser, J.
L. (2019). Exploiting Honokiol‐induced ER stress CHOP activation inhibits
apoptosis via endoplasmic reticulum stress” sections. ST scientifically
the growth and metastasis of melanoma by suppressing the MITF and β‐
edited the manuscript and wrote the abstract and “Melanoma and catenin pathways. Cancer Letters, 442, 113–125.
autophagy” section. ZA wrote the “Anoikis”, “Necroptosis” and Cohen, G. M. (1997). Caspases: The executioners of apoptosis. Biochemical
“Ferroptosis” sections. SR wrote the “Endoplasmic reticulum stress” Journal, 326(1), 1–16.
Cruz, S. A., Qin, Z., Stewart, A. F., & Chen, H.‐H. (2018). Dabrafenib, an
and “Induction of autophagy and apoptosis via endoplasmic reticulum
inhibitor of RIP3 kinase‐dependent necroptosis, reduces ischemic
stress” sections. MK wrote the “Melanoma and anoikis” section. brain injury. Neural Regeneration Research, 13(2), 252.
ASHRAFIZADEH ET AL. | 19477

Cuervo, A. M., & Wong, E. (2014). Chaperone‐mediated autophagy: Roles glioma cells. Biomedicine & Pharmacotherapy, 93, 969–975. https://doi.
in disease and aging. Cell Research, 24(1), 92–104. org/10.1016/j.biopha.2017.07.010
Dixon, S. J., Lemberg, K. M., Lamprecht, M. R., Skouta, R., Zaitsev, E. M., Gupta, P., Gupta, N., Fofaria, N. M., Ranjan, A., & Srivastava, S. K. (2019).
Gleason, C. E., & Yang, W. S. (2012). Ferroptosis: An iron‐dependent HER2‐mediated GLI2 stabilization promotes anoikis resistance and
form of nonapoptotic cell death. Cell, 149(5), 1060–1072. metastasis of breast cancer cells. Cancer Letters, 442, 68–81.
Dolma, S., Lessnick, S. L., Hahn, W. C., & Stockwell, B. R. (2003). Hadifar, S., Behrouzi, A., Fateh, A., Khatami, S., Rahimi Jamnani, F., Siadat,
Identification of genotype‐selective antitumor agents using synthetic S. D., & Vaziri, F. (2018). Comparative study of interruption of
lethal chemical screening in engineered human tumor cells. Cancer signaling pathways in lung epithelial cell by two different Mycobacter-
Cell, 3(3), 285–296. ium tuberculosis lineages. Journal of Cellular Physiology., 234(4), 4739–
Donato, A. L., Huang, Q., Liu, X., Li, F., Zimmerman, M. A., & Li, C.‐Y. (2014). 4753.
Caspase 3 promotes surviving melanoma tumor cell growth after Hammerová, J., Uldrijan, S., Táborská, E., Vaculová, A. H., & Slaninová, I.
cytotoxic therapy. Journal of Investigative Dermatology, 134(6), 1686– (2012). Necroptosis modulated by autophagy is a predominant form
1692. of melanoma cell death induced by sanguilutine. Biol Chem, 393, 647–
Faghihi, F., Mirzaei, E., Ai, J., Lotfi, A., Sayahpour, F. A., Barough, S. E., & 658.
Joghataei, M. T. (2016). Differentiation potential of human chorion‐ Hasnat, M. A., Pervin, M., Lim, J. H., & Lim, B. O. (2015). Apigenin attenuates
derived mesenchymal stem cells into motor neuron‐like cells in two‐ melanoma cell migration by inducing anoikis through integrin and focal
and three‐dimensional culture systems. Molecular Neurobiology, 53(3), adhesion kinase inhibition. Molecules, 20 (12), 21157–21166.
1862–1872. He, J., Dong, L., Xu, K., Qian, Y., Wang, C., Sha, Y., & Song, Y. (2018).
Fitzwalter, B. E., Towers, C. G., Sullivan, K. D., Andrysik, Z., Hoh, M., Mechano growth factor E peptide inhibits invasion of melanoma cells
Ludwig, M., & Morgan, M. J. (2018). Autophagy inhibition mediates and up‐regulates CHOP expression via endoplasmic reticulum stress.
apoptosis sensitization in cancer therapy by relieving FOXO3a Biotechnology Letters, 40 (1), 205–213.
turnover. Developmental Cell, 44(5), 555–565. e553 Heo, J. R., Kim, S. M., Hwang, K. A., Kang, J. H., & Choi, K. C. (2018).
Fofaria, N. M., & Srivastava, S. K. (2014). Critical role of STAT3 in Resveratrol induced reactive oxygen species and endoplasmic reticu-
melanoma metastasis through anoikis resistance. Oncotarget, 5(16), lum stress‑mediated apoptosis, and cell cycle arrest in the A375SM
7051. malignant melanoma cell line. International Journal of Molecular
Frisch, S. M., & Screaton, R. A. (2001). Anoikis mechanisms. Current Medicine., 42, 1427–1435.
Opinion in Cell Biology, 13(5), 555–562. Heo, J.‐R., Lee, G.‐A., Kim, G.‐S., Hwang, K.‐A., & Choi, K.‐C. (2018).
Gallagher, R. P. (2005). Sunscreens in melanoma and skin cancer Phytochemical‐induced reactive oxygen species and endoplasmic
prevention. Canadian Medical Association Journal/Journal de l'Associa- reticulum stress‐mediated apoptosis and differentiation in malignant
tion Medicale Canadienne, 173(3), 244–245. melanoma cells. Phytomedicine, 39, 100–110.
Gallazzini, M., & Pallet, N. (2018). Endoplasmic reticulum stress and Hollien, J., & Weissman, J. S. (2006). Decay of endoplasmic reticulum‐
kidney dysfunction. Biology of the Cell, 110 (9), 205–216. localized mRNAs during the unfolded protein response. Science,
Galluzzi, L., López‐Soto, A., Kumar, S., & Kroemer, G. (2016). Caspases 313(5783), 104–107.
connect cell‐death signaling to organismal homeostasis. Immunity, Hurle, J. M. (2014). Regenerative versus destructive cell death in
44(2), 221–231. developing systems and tissue homeostasis. Bioessays, 36
Galluzzi, L., Kepp, O., Chan, F. K.‐M., & Kroemer, G. (2017). Necroptosis: Kakavandi, E., Shahbahrami, R., Goudarzi, H., Eslami, G., & Faghihloo, E.
Mechanisms and relevance to disease. Annual Review of Pathology: (2018). Anoikis resistance and oncoviruses. Journal of Cellular
Mechanisms of Disease, 12, 103–130. Biochemistry, 119(3), 2484–2491.
Galluzzi, L., Bravo‐San Pedro, J., Vitale, I., Aaronson, S., Abrams, J., Adam, Kimmelman, A. C., & White, E. (2017). Autophagy and tumor metabolism.
D., & Annicchiarico‐Petruzzelli, M. (2015). Essential versus accessory Cell Metabolism, 25(5), 1037–1043.
aspects of cell death: Recommendations of the NCCD 2015. Cell Klionsky, D. J., & Codogno, P. (2013). The mechanism and physiological
Death and Differentiation, 22(1), 58–73. function of macroautophagy. Journal of Innate Immunity, 5(5), 427–433.
Galluzzi, L., Vitale, I., Aaronson, S. A., Abrams, J. M., Adam, D., Agostinis, P., Klionsky, D. J., Baehrecke, E. H., Brumell, J. H., Chu, C. T., Codogno, P.,
& Andrews, D. W. (2018). Molecular mechanisms of cell death: Cuervo, A. M., & Eskelinen, E.‐L. (2011). A comprehensive glossary of
Recommendations of the Nomenclature Committee on Cell Death autophagy‐related molecules and processes. Autophagy, 7(11), 1273–
2018. Cell Death & Differentiation, 1 1294.
Gao, M., Monian, P., Pan, Q., Zhang, W., Xiang, J., & Jiang, X. (2016). Kong, Q., Lv, J., Yan, S., Chang, K.‐J., & Wang, G. (2018). A novel
Ferroptosis is an autophagic cell death process. Cell Research, 26(9), naphthyridine derivative, 3u, induces necroptosis at low concentra-
1021–1032. tions and apoptosis at high concentrations in human melanoma A375
Geserick, P., Wang, J., Schilling, R., Horn, S., Harris, P., Bertin, J., & cells. International Journal of Molecular Sciences, 19(10), 2975.
Leverkus, M. (2015). Absence of RIPK3 predicts necroptosis Kuwana, T., Bouchier‐Hayes, L., Chipuk, J. E., Bonzon, C., Sullivan, B. A.,
resistance in malignant melanoma. Cell Death & Disease, 6(9), e1884. Green, D. R., & Newmeyer, D. D. (2005). BH3 domains of BH3‐only
Gilmore, A. (2005). Anoikis, Cell Death and Differentiation 12, 1473–1477). proteins differentially regulate Bax‐mediated mitochondrial mem-
New York brane permeabilization both directly and indirectly. Molecular Cell,
Goldstein, N. B., Johannes, W. U., Gadeliya, A. V., Green, M. R., Fujita, M., 17(4), 525–535.
Norris, D. A., & Shellman, Y. G. (2009). Active N‐Ras and B‐Raf inhibit Latunde‐Dada, G. O. (2017). Ferroptosis: Role of lipid peroxidation, iron
anoikis by downregulating Bim expression in melanocytic cells. Journal and ferritinophagy. Biochimica et Biophysica Acta (BBA)‐General
of Investigative Dermatology, 129(2), 432–437. Subjects, 1861(8), 1893–1900.
Goundiam, O., Nagel, M. D., & Vayssade, M. (2012). Akt and RhoA Li, S., Ning, L.‐G., Lou, X.‐H., & Xu, G.‐Q. (2018). Necroptosis in
inhibition promotes anoikis of aggregated B16F10 melanoma cells. inflammatory bowel disease and other intestinal diseases. World
Cell Biology International, 36(3), 311–319. Journal of Clinical Cases, 6(14), 745–752.
Grossmann, J. (2002). Molecular mechanisms of “detachment‐induced Luan, W., Qian, Y., Ni, X., Chanda, T. K., Xia, Y., Wang, J., & Xu, B. (2017).
apoptosis—Anoikis”. Apoptosis, 7(3), 247–260. Polygonatum odoratum lectin promotes BECN1 expression and
Gu, J., Lu, Z., Ji, C., Chen, Y., Liu, Y., Lei, Z., & Li, X. (2017). Melatonin induces autophagy in malignant melanoma by regulation of
inhibits proliferation and invasion via repression of miRNA‐155 in mir1290. OncoTargets and Therapy, 10 , 4569–4577.
19478 | ASHRAFIZADEH ET AL.

Luo, M., Wu, L., Zhang, K., Wang, H., Zhang, T., Gutierrez, L., & Gao, T. Shang, Y.‐Y., Yao, M., & Zhou, Z.‐W. (2017). Alisertib promotes apoptosis
(2018). miR‐137 regulates ferroptosis by targeting glutamine trans- and autophagy in melanoma through p38 MAPK‐mediated aurora a
porter SLC1A5 in melanoma. Cell Death & Differentiation, 1 signaling. Oncotarget, 8(63), 107076–107088.
Mawji, I. A., Simpson, C. D., Gronda, M., Williams, M. A., Hurren, R., Shen, J., Chen, X., Hendershot, L., & Prywes, R. (2002). ER stress
Henderson, C. J., & Schimmer, A. D. (2007). A chemical screen regulation of ATF6 localization by dissociation of BiP/GRP78 binding
identifies anisomycin as an anoikis sensitizer that functions by and unmasking of Golgi localization signals. Developmental Cell, 3(1),
decreasing FLIP protein synthesis. Cancer Research, 67(17), 8307– 99–111.
8315. Tarver, T. (2012). Cancer facts & figures 2012. American cancer society
Meares, G. P., Hughes, K. J., Naatz, A., Papa, F. R., Urano, F., Hansen, P. A., (ACS) Atlanta, GA: American Cancer Society. 2012. 66 p., pdf.
& Corbett, J. A. (2011). IRE1‐dependent activation of AMPK in Available from: Taylor & Francis
response to nitric oxide. Molecular and Cellular Biology, 31(21), 4286– Tavakol, S. (2014). Acidic pH derived from cancer cells may induce failed
4297. reprogramming of normal differentiated cells adjacent tumor cells and
Menzies, F. M., Fleming, A., & Rubinsztein, D. C. (2015). Compromised turn them into cancer cells. Medical Hypotheses, 83(6), 668–672.
autophagy and neurodegenerative diseases. Nature Reviews Neu- Tavakol, S., Jalili‐Firoozinezhad, S., Mashinchian, O., & Mahmoudi, M. (2016).
roscience, 16(6), 345–357. Bioinspired nanotechnologies for skin regeneration, Nanoscience in
Mgrditchian, T., Arakelian, T., Paggetti, J., Noman, M. Z., Viry, E., Moussay, Dermatology (337–352). Elsevier.
E., & Buart, S. (2017). Targeting autophagy inhibits melanoma growth Tavakol, S., Zare, S., Hoveizi, E., Tavakol, B., & Rezayat, S. M. (2019). The
by enhancing NK cells infiltration in a CCL5‐dependent manner. impact of the particle size of curcumin nanocarriers and the ethanol
Proceedings of the National Academy of Sciences, 114(44), E9271– on beta_1‐integrin overexpression in fibroblasts: A regenerative
E9279. pharmaceutical approach in skin repair and anti‐aging formulations.
Mohammadinejad, R., Ahmadi, Z., Tavakol, S., & Ashrafizadeh, M. (2019). DARU Journal of Pharmaceutical Sciences, 1–10.
Berberine as a potential autophagy modulator. Journal of Cellular Tavakol, S., Mousavi, S. M. M., Tavakol, B., Hoveizi, E., Ai, J., & Sorkhabadi,
Physiology. S. M. R. (2017). Mechano‐transduction signals derived from self‐
Mohammadinejad, R., Moosavi, M. A., Tavakol, S., Vardar, D. Ö., Hosseini, assembling peptide nanofibers containing long motif of laminin
A., Rahmati, M., & Klionsky, D. J. (2018). Necrotic, apoptotic and influence neurogenesis in in‐vitro and in‐vivo. Molecular Neurobiology,
autophagic cell fates triggered by nanoparticles. Autophagy, 15(1), 4– 54(4), 2483–2496.
33. Taylor, R. C., Cullen, S. P., & Martin, S. J. (2008). Apoptosis: Controlled
Mohammadinejad, R., Dadashzadeh, A., Moghassemi, S., Ashrafizadeh, M., demolition at the cellular level. Nature Reviews Molecular Cell Biology,
Dehshahri, A., Pardakhty, A., & Mandegary, A. (2019). Shedding light 9(3), 231–241.
on gene therapy: Carbon dots for the minimally invasive image‐guided Terasaki, M., Iida, T., Kikuchi, F., Tamura, K., Endo, T., Kuramitsu, Y., &
delivery of plasmids and noncoding RNAs. Journal of Advanced Mutoh, M. (2018). Fucoxanthin potentiates anoikis in colon mucosa
Research, 18, 81–93. and prevents carcinogenesis in AOM/DSS model mice. The Journal of
Mollazadeh, H., Atkin, S. L., Butler, A. E., Ruscica, M., Sirtori, C. R., & Nutritional Biochemistry, 64, 198–205.
Sahebkar, A. (2018). The effect of statin therapy on endoplasmic Thorburn, J., Andrysik, Z., Staskiewicz, L., Gump, J., Maycotte, P., Oberst,
reticulum stress. Pharmacological Research, 137, 150–158. A., & Thorburn, A. (2014). Autophagy controls the kinetics and extent
Nikseresht, S., Khodagholi, F., & Ahmadiani, A. (2018). Protective effects of mitochondrial apoptosis by regulating PUMA levels. Cell Reports,
of ex‐527 on cerebral ischemia–reperfusion injury through necropto- 7(1), 45–52.
sis signaling pathway attenuation. Journal of Cellular Physiology., 234, Toricelli, M., Melo, F. H., Peres, G. B., Silva, D. C., & Jasiulionis, M. G. (2013).
1816–1826. Timp1 interacts with beta‐1 integrin and CD63 along melanoma genesis
Nyfeler, B., & Eng, C. H. (2016). Revisiting autophagy addiction of tumor and confers anoikis resistance by activating PI3‐K signaling pathway
cells. Autophagy, 12(7), 1206–1207. independently of Akt phosphorylation. Molecular Cancer, 12(1), 1095.
Ofengeim, D., & Yuan, J. (2013). Regulation of RIP1 kinase signalling at the Tseng, T., Uen, W., Tseng, J., & Lee, S. (2017). Enhanced chemosensitiza-
crossroads of inflammation and cell death. Nature Reviews Molecular tion of anoikis‐resistant melanoma cells through syndecan‐2 upregu-
Cell Biology, 14(11), 727–736. lation upon anchorage independency. Oncotarget, 8(37), 61528.
Pasparakis, M., & Vandenabeele, P. (2015). Necroptosis and its role in Villalobos‐Labra, R., Subiabre, M., Toledo, F., Pardo, F., & Sobrevia, L.
inflammation. Nature, 517(7534), 311–320. (2018). Endoplasmic reticulum stress and development of insulin
Rabiee, S., Tavakol, S., Barati, M., & Joghataei, M. T. (2018). Autophagic, resistance in adipose, skeletal, liver, and foetoplacental tissue in
apoptotic, and necrotic cancer cell fates triggered by acidic pH diabesity. Molecular Aspects of Medicine., 30080–30083.
microenvironment. Journal of Cellular Physiology, 234, 12061– Walter, P., & Ron, D. (2011). The unfolded protein response: From stress
12069. pathway to homeostatic regulation. Science, 334(6059), 1081–1086.
Rebecca, V., & Amaravadi, R. (2016). Emerging strategies to effectively White, E. (2016). Autophagy and p53. Cold Spring Harbor Perspectives in
target autophagy in cancer. Oncogene, 35(1), 1–11. Medicine, 6(4), a026120.
Roumane, A., Berthenet, K., El Fassi, C., & Ichim, G. (2018). Caspase‐ Wu, J.‐C., Tsai, H.‐E., Liu, G.‐S., Wu, C.‐S., & Tai, M.‐H. (2018). Autophagic
independent cell death does not elicit a proliferative response in cell death participates in POMC‐induced melanoma suppression. Cell
melanoma cancer cells. BMC Cell Biology, 19(1), 11. Death Discovery, 5(1), 11.
Ryabaya, O., Prokofieva, A., Khochenkov, D., Abramov, I., Zasedatelev, A., Xiao, Y., Diao, Q., Liang, Y., Peng, Y., & Zeng, K. (2017). MicroRNA‐24‐1‐5p
& Stepanova, E. (2018). Inhibition of endoplasmic reticulum stress‐ promotes malignant melanoma cell autophagy and apoptosis via
induced autophagy sensitizes melanoma cells to temozolomide regulating ubiquitin D. Molecular Medicine Reports, 16(6), 8448–8454.
treatment. Oncology Reports, 40 (1), 385–394. Xie, Y., Hou, W., Song, X., Yu, Y., Huang, J., Sun, X., & Tang, D. (2016).
Shakeri, A., Cicero, A. F., Panahi, Y., Mohajeri, M., & Sahebkar, A. (2018). Ferroptosis: Process and function. Cell Death and Differentiation,
Curcumin: A naturally occurring autophagy modulator. Journal of 23(3), 369–379.
Cellular Physiology., 234, 5643–5654. Yang, T.‐Y., Wu, Y.‐J., Chang, C.‐I., Chiu, C.‐C., & Wu, M.‐L. (2018). The
Shan, B., Pan, H., Najafov, A., & Yuan, J. (2018). Necroptosis in effect of Bornyl cis‐4‐hydroxycinnamate on melanoma cell apoptosis
development and diseases. Genes & development, 32(5‐6), 327– is associated with mitochondrial dysfunction and endoplasmic
340. reticulum stress. International Journal of Molecular Sciences, 19(5).
ASHRAFIZADEH ET AL. | 19479

Yang, W. S., SriRamaratnam, R., Welsch, M. E., Shimada, K., Skouta, R., Zheng, Y., Wang, K., Wu, Y., Chen, Y., Chen, X., Hu, C. W., & Hu, F. (2018).
Viswanathan, V. S., & Clish, C. B. (2014). Regulation of ferroptotic Pinocembrin induces ER stress mediated apoptosis and suppresses
cancer cell death by GPX4. Cell, 156(1‐2), 317–331. autophagy in melanoma cells. Cancer Letters, 431, 31–42.
Yoshida, H., Okada, T., Haze, K., Yanagi, H., Yura, T., Negishi, M., & Mori, Zhu, Z., Sanchez‐Sweatman, O., Huang, X., Wiltrout, R., Khokha, R., Zhao,
K. (2000). ATF6 activated by proteolysis binds in the presence of Q., & Gorelik, E. (2001). Anoikis and metastatic potential of cloudman
NF‐Y (CBF) directly to the cis‐acting element responsible for the S91 melanoma cells. Cancer Research, 61(4), 1707–1716.
mammalian unfolded protein response. Molecular and Cellular
Biology, 20 (18), 6755–6767.
Zalckvar, E., Berissi, H., Mizrachy, L., Idelchuk, Y., Koren, I., Eisenstein, M., SU PP ORT IN G IN FOR MATI O N
& Kimchi, A. (2009). DAP‐kinase‐mediated phosphorylation on the
BH3 domain of beclin 1 promotes dissociation of beclin 1 from Bcl‐XL Additional supporting information may be found online in the
and induction of autophagy. EMBO Reports, 10 (3), 285–292. Supporting Information section at the end of the article.
Zendedel, A., Nobakht, M., Bakhtiyari, M., Beyer, C., Kipp, M., Baazm, M., &
Joghataie, M. T. (2012). Stromal cell‐derived factor‐1 alpha (SDF‐1α)
improves neural recovery after spinal cord contusion in rats. Brain
Research, 1473, 214–226. How to cite this article: Ashrafizadeh M, Mohammadinejad R,
Zhang, K., Wu, L., Zhang, P., Luo, M., Du, J., Gao, T., & Yang, Y. (2018). miR‐9 Tavakol S, Ahmadi Z, Roomiani S, Katebi M. Autophagy, anoikis,
regulates ferroptosis by targeting glutamic‐oxaloacetic transaminase
ferroptosis, necroptosis, and endoplasmic reticulum stress:
GOT1 in melanoma. Molecular Carcinogenesis, 57(11), 1566–1576.
Zhang, Y., Yang, M., Ji, Q., Fan, D., Peng, H., Yang, C., & Zhou, Y. (2011). Anoikis
Potential applications in melanoma therapy. J Cell Physiol. 2019;
induction and metastasis suppression by a new integrin αvβ3 inhibitor in 234:19471–19479. https://doi.org/10.1002/jcp.28740
human melanoma cell line M21. Investigational New Drugs, 29(4), 666–673.

You might also like