You are on page 1of 12

Biomedicine & Pharmacotherapy 149 (2022) 112827

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Advances in indole-containing alkaloids as potential anticancer agents by


regulating autophagy
Meng-Lan Luo a, Wei Huang a, Hong-Ping Zhu a, b, Cheng Peng a, Qian Zhao a, *, Bo Han a, *
a
State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu
University of Traditional Chinese Medicine, Chengdu, China
b
Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China

A R T I C L E I N F O A B S T R A C T

Keywords: Cancer is a leading cause of death worldwide, and cancer development is often associated with disturbances in
Indole the autophagy process. Autophagy is a catabolic process involved in many physiological processes, crucial for cell
Autophagy-related growth and survival. It is an intracellular lysosomal/vacuolar degradation system. In this system, inner cyto­
Cancer therapy
plasmic cell membrane is degraded by lysosomal hydrolases, and the products are released back into the cyto­
Signaling pathway
plasm. Indole alkaloids are natural products extensively found in nature and have been proven to possess various
Cell survival
Cell death pharmacological activities. In recent years, pharmacological studies have demonstrated another potential of
indole alkaloids, autophagy regulation. The regulation may contribute to the efficacy of indole alkaloids in
preventing and treating cancer. This review summarizes the current understanding of indole alkaloids’ effect on
tumor cells and autophagy. Then, we focus on mechanisms by which indole alkaloids can target the autophagy
process associated with cancer, including the PI3K/Akt/mTOR signaling pathway, MAPK signaling pathway,
ROS signaling pathway, Beclin-1, and so on. Literature has been surveyed primarily from 2009 to Nov. 2021, and
some semisynthetic or fully synthetic indole derivatives are also discussed.

1. Introduction compounds in traditional medicine remains a popular and fruitful


approach [3–6]. Numerous natural products have been isolated from
Cancer is a leading cause of death and a significant barrier to plants and proved to have high cytotoxicity and antitumor activity [7].
increasing life expectancy in every country in the world. Statistical data Indole alkaloids, one of the most abundant and complex alkaloids,
published by the World Health Organization International Agency for ubiquitously exist in natural products. Various indole-containing natural
Cancer Research have recorded 19.3 million new cancer cases world­ compounds have already been proven to exhibit a broad of pharmaco­
wide and nearly 10 million cancer deaths [1]. Despite the immense logical activities, such as antibacterial, antifungal, antimalarial, anti-
treatments and techniques available for cancer treatment and preven­ inflammatory, anti-HIV, anti-plasmodial, and especially antitumor ac­
tion, many drugs currently have high side effects and multidrug resis­ tivities [8–19]. The prominent role of indole alkaloids’ biological ac­
tance in clinical practice [2]. In this context, developing safer and more tivity over the past decades may reflect their increasing utility for
reliable alternatives is essential. Identifying plant-based lead synthesizing pharmaceuticals and bioactive compounds. In addition,

Abbreviations: PI3K, Phosphatidylinositol 3-kinase; Akt, Protein kinase B; mTOR, Mammalian target of rapamycin; MAPK, Mitogen-activated protein kinase 1;
ROS, Reactive oxygen species; NSCLC, Non-small cell lung cancer; CMA, Chaperone-mediated autophagy; ATP, Adenosine triphosphate; ULK1, Unc-51-like kinase 1;
FIP200, Family kinase-interacting protein 200; Atg, Autophagy-related gene; mTORC1, Rapamycin complex 1; PE, Phosphatidyl Ethanolamine, SNAP, Synaptosomal-
associated protein; VAMP, Vesicle-associated membrane protein; AMPK, 5’-AMP-activated protein kinase; Bcl-2, B-cell lymphoma 2; MNZQ, Muniziqi granule; VEGF,
Vascular endothelial growth factor; I3C, Indole-3-carbinol; ERK, Extracellular signal-related kinases; LAMP, Lysosomal-associated membrane protein 1; JNK, c-Jun N-
terminal kinase; 11-MT, 11-methoxytabersonine; CAA, Calothrixin A; OSCC, Oral squamous cell carcinoma; ER, Endoplasmic reticulum; VPS, Vacuolar protein
sorting; BH3, Bcl-2-homology-3 domain; Hsp, Heat shock protein; NADPH, Nicotinamide adenine dinucleotide phosphate; FoxO3, Forkhead box protein O3; R-scy,
Reduced scytonemin; UBA, Ubiquitin-associated; LIR, LC3-interacting region; PB1, N-terminal phox-BEM1; PPARA, Peroxisome proliferator-activated receptor alpha;
CPT, Carnitine palmityl transferase; NAFLD, Non-alcoholic fatty liver disease.
* Corresponding authors.
E-mail addresses: zhaoqian@cdutcm.edu.cn (Q. Zhao), hanbo@cdutcm.edu.cn (B. Han).

https://doi.org/10.1016/j.biopha.2022.112827
Received 27 January 2022; Received in revised form 3 March 2022; Accepted 14 March 2022
Available online 19 March 2022
0753-3322/© 2022 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

many indole alkaloids and their derivatives are commonly applied in (CMA) [37–39]. The term “autophagy” is usually referred to macro­
clinical practice to treat various types of cancer, such as acute leukemia, autophagy, which is the most extensively studied one. In eukaryotic
malignant lymphoma, small-cell lung cancer, and breast cancer [20–27] cells, autophagy is caused by a series of factors, such as reduced insulin
(Fig. 1). Thus, the indole framework is a fascinating and privileged levels, amino acid starvation, decreased ATP levels, and hypoxia. The
scaffold for the development of novel drugs. Moreover, in the contin­ process of autophagy consists of five steps: initiation, nucleation,
uous research of our group on bioactive indole derivatives, we have also maturation, fusion, and degradation. The initiation of autophagy is
explored a series of indole compounds with high biological activity controlled by Unc-51-like kinase 1 (ULK1) complex, which is comprised
[28–34]. Therefore, these chemical entities can be used as lead com­ of the serine/threonine-protein kinase ULK1, family kinase-interacting
pounds in new-drug development. Actually, increasing evidence in­ protein 200 (FIP200), autophagy-related gene 13 (Atg 13) [40]. Upon
dicates that bioactive indole alkaloids exert their antitumor activity autophagy stimuli, such as amino acid starvation and decreased insulin
through autophagy regulation. levels, rapamycin complex 1 (mTORC1) is inhibited, which can lead to
Autophagy, a catabolic process, plays an essential role in regulating activation of the ULK1 complex [41]. In the nucleation step of auto­
cellular homeostasis in physiological and pathophysiological conditions phagy, the ULK1 complex phosphorylates and activates the
[35,36]. To date, three primary types of autophagy have been described: Beclin-1-class III phosphatidylinositol 3 Kinase (PI3K) complex. This
macroautophagy, microautophagy, and chaperone-mediated autophagy complex is comprised of Beclin-1, vacuolar protein sorting (VPS34, a

Fig. 1. The chemical structures of anti-cancer indole alkaloids and indole derivatives. (1). Vinblastine is an anti-cancer drug extracted from Catharanthus roseus. This
drug is given to treat Hodgkin lymphoma, non-small cell lung cancer (NSCLC), head and neck cancer. (2). Vincristine is an indole alkaloid isolated from leaves of the
Madagascar periwinkle plant, Catharanthus roseus. Vincristine is used in the treatment of low-grade gliomas and anaplastic oligodendrogliomas. (3). Vinorelbine is a
semi-synthetic derivative of vinblastine and is used in NSCLC, breast, and ovarian cancers. (4). Rucaparib camsylate is a poly ADP-ribose polymerase inhibitor and is
the world’s first third-line treatment for ovarian cancer. (5). Alectinib hydrochloride is an anaplastic lymphoma kinase inhibitor and is used in the treatment of
recurrent non-small cell lung cancer. (6) Osimertinib is the first and only NSCLC drug approved for the EGFR T790M mutation. (7). Anlotinib Dihydrochloride is a
small molecule multi-target tyrosine kinase inhibitor used in the treatment of alveolar soft tissue sarcoma. (8). Panobinostat is the first histone deacetylase inhibitor
for the treatment of multiple myeloma.

2
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

class III phosphatidylinositol 3-kinase), and other proteins [42]. Two signal-transduction enzyme in this signaling pathway and is a lipid ki­
ubiquitin-like binding reaction systems are involved in the formation of nase that catalyzes the phosphorylation of the phosphatidylinositol at
autophagosomes: 1) LC3-Phosphatidyl Ethanolamine (PE) binding re­ D3 in the cytoplasm [47,48].
action system, 2) Atg12-Atg5 binding reaction system [43]. In the fusion As a crucial downstream target kinase of PI3K, Akt is a serine/thre­
process, the formation of the conjugates promotes the fusion of auto­ onine kinase, also known as Protein kinase B. Two activated sites,
phagosomes and lysosomes. The conjugates include namely, Thr308 and Ser473, can fully activate Akt together [49]. mTOR
synaptosomal-associated protein (SNAP)29, lysosomal is a central regulator of autophagy and a serine/threonine kinase
vesicle-associated membrane protein (VAMP)7/VAMP8, and SNARE belonging to the phosphoinositol 3-kinase-related protein kinase family
[44] (Fig. 2). LC3-II protein is often used as an autophagosome marker. [50]. It comprises two distinct signaling complexes named mTORC1 and
Several signaling pathways have been implicated in autophagy regula­ mTORC2. However, as the main effector of the PIK downstream
tion, including pathways mediated by PI3K/Akt/mammalian target of pathway, mTORC1 plays a key role in direct autophagy regulation
rapamycin (mTOR), 5’-AMP-activated protein kinase (AMPK)/mTOR, [51–53]. mTORC1 can directly inhibit the activity of ULK1 by phos­
Mitogen-activated protein kinase (MAPK), Beclin-1, Bcl-2, p53, and p38 phorylating the complex of ULK1, which constitutes ULK1, Atg13,
(Fig. 3) [45]. FIP200, and Atg101. Under nutrient-poor conditions, dephosphoryla­
The antitumor activity of indole alkaloids has been extensively tion of mTORC1 results in the departure from the ULK1 complex, which
studied in the field of drug development. A mechanistic study has allows the activation of ULK1. Concomitantly, ULK1 undergoes auto­
revealed that indole alkaloids regulate autophagy participating in the phosphorylation and dephosphorylates ATG 13 and FIP200, which are
PI3K/Akt/mTOR signaling pathway, MAPK signaling pathway, ROS involved in autophagy initiation [54–56]. Many indole alkaloids and
signaling pathway, and Beclin-1, leading to antitumor activity. Despite their derivatives can more accurately and specifically regulate the
this progress, no comprehensive review has been devoted to this topic. autophagy activity of tumor cells by targeting
In this review, we systemically presented recent advances in indole al­ PI3K/Akt/mTOR-mediated autophagy to exhibit antitumor activity.
kaloids as anticancer agents by regulating autophagy categorized as β-Carboline alkaloids are a remarkable family of natural indole-
follows: (1) targeting the PI3K/Akt/mTOR signaling pathway, (2) tar­ containing compounds, and they are widely distributed in many kinds
geting the MAPK signaling pathway, (3) targeting Beclin-1, (4) targeting of medicinal plants. Recently, these alkaloids have been the focus of
the ROS signaling pathway, (5) targeting P62/SQSTM1, and (6) tar­ interest, due to their diverse biological activities, such as antitumor,
geting miscellaneous pathways. Literature has been surveyed primarily antiviral, antiparasitic, and anxiolytic. Abe and his coworkers [57]
from 2009 to Nov. 2021, and some semisynthetic or fully synthetic demonstrated that harmol induces autophagy and subsequently induces
indole derivatives are also discussed. A list of chemical structures indole- apoptotic cell death in U251MG human glioma cells by reducing phos­
containing molecules is presented in Fig. 4. phorylation of Akt, mTOR, and its downstream targets. Notably, here
harmol-induced autophagy is a pro-death mechanism by suppressing
2. Targeting the PI3K/Akt/mTOR signaling pathway survivin protein expression. Compound Muniziqi granule (MNZQ) is a
traditional Chinese multi-component herbal preparation and can treat
The PI3K/Akt/mTOR signaling pathway extensively exists in human endocrine disorders caused by acne, chloasma, dysmenorrhea, meno­
cells and plays an important role in maintaining normal cellular activity. pausal syndrome, and melanoma [58]. Harmine is one of the most
It is also a critical pathway for autophagy regulation [46]. The potent anti-melanoma agents extracted from MNZQ. Harmine has been
PI3K/Akt/mTOR signaling pathway comprises three important reported to induce the formation of autophagosomes via the increase of
signaling molecules: PI3K, Akt, and mTOR. PI3K is the first LC3-II in B16 cells, which shows that it is an autophagy inducer [59]. In

Fig. 2. The process of autophagy. Under the amino acid or nutrient starvation conditions, the ULK1 complex (ULK1, Atg13, FIP200, and Atg101) gets activated by
coordinated inputs from the mTORC1, which allows the initiation of autophagy. The ULK1 complex phosphorylates and activates the Beclin-1/Vps34 complex in the
nucleation step of autophagy. In the process of vesicle expansion and completion, two ubiquitin-like binding reaction systems are involved in forming autopha­
gosomes, namely the LC3-PE binding reaction system and the Atg12-Atg5 binding reaction system. The conjugates included SNAP29, VAMP7/VAMP8, and SNARE
play a vital role in the fusion process.

3
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

Fig. 3. Targeting signaling cascades regulating autophagy. Various pathways regulate autophagy. mTOR kinase is an important regulatory molecule that induces
autophagy. Activated mTOR inhibits autophagy, while negative regulation of mTOR promotes autophagy. AMPK and P53 activate autophagy directly via phos­
phorylating ULK1 or indirectly via phosphorylating TSC1/2, which suppresses mTORC1 activity. Akt and MAPK activate mTORC1 to inhibit autophagy. ROS ac­
tivates autophagy via multiple pathways, including NOX4 activation, which promotes the PERK/eIF2α/ATF4 pathway.

addition, the result indicated that harmine-mediated autophagy was candidate drug for cancer treatment.
activated by inhibiting the Akt/mTOR pathway and partly activating the Bisleuconothine A, a novel bisindole alkaloid isolated from Leuco­
ERK1/2 pathway. The cell death induced by harmine suggests it could notis griffithii, has higher inhibitory activity in MCF7 cells and A549
be a promising therapeutic agent for the treatment of melanoma. Af­ cells, and the effect is associated with autophagy-mediated by disturbing
terward, Li et al. [60] proved that harmine significantly upregulates the PI3K/Akt/mTOR signaling pathway. Notably, bisleuconothine A
autophagy and apoptosis activity in human gastric cancer cells, which is induces autophagosome formation but inhibits autophagic flux [64].
involved in cell death. This induction is mediated by the inhibition of the Staurosporine is a natural product with a bisindole structure extracted
Akt/mTOR/p70S6K signaling pathway and the AMPK signaling from the bacterium Streptomyces staurosporeus. In HepG2 cancer cells,
pathway, which is related to the expression of Beclin-1, LC3-II, and p62 staurosporine significantly induces autophagy by inhibiting the
[60]. PI3K/Akt/mTOR signaling pathway, which allows the cytotoxicity
Fascaplysin, a marine natural active product, has wide-ranging bio­ response [65]. The effect provides a novel mechanism by which staur­
activities, such as antibacterial, antifungal, antiviral, antimalarial, and osporine exerts its therapeutic effects. Indole-3-carbinol (I3C) is an
anticholinesterase [61]. Kumar’s group found that fascaplysin induces indole alkaloid extracted from cruciferous vegetables. It exerts anti­
cell death in HL-60 cells attributed to the cooperative interaction be­ cancer, antioxidant, and anti-inflammatory effects. Jiang and his co­
tween apoptosis and autophagy. The effect on autophagy is activated by workers found that I3C exerts protective effects on hyperlipidemia
inhibiting the PI3K/Akt/mTOR signaling pathway, resulting in zebrafish larvae by promoting autophagy. Moreover, the induction of
increased expression of LC3-II, ATG7, and Beclin-1 [62]. This suggests autophagy has been shown by the enhancement in LC3-II and Beclin-1,
that the synergy of autophagy and apoptosis plays a crucial role in the as well as the reduction in P62, Bcl-2, Akt, and mTOR, through the
antitumor effect of fascaplysin. Sharma et al. [63] synthesized a series of PI3K/Akt/mTOR pathway [66].
marine sponge alkaloid fascaplysin analogs. One of the compounds,
fascaplysin chloride, inhibits vascular endothelial growth factor 3. Targeting the MAPK signaling pathway
(VEGF)-mediated angiogenesis and induces autophagy and apoptosis
through the interference with the PI3K/Akt/mTOR signaling pathway in MAPKs are signaling components that are essential for converting
MDAMB-231 cells [63]. This finding suggests that fascaplysin derivative extracellular stimuli into a wide array of cellular processes [67]. MAPK
has antiangiogenesis and antiproliferative potential. The results of both signaling pathway is also one of the most important transduction
studies show that fascaplysin and its derivatives could be a good pathways that mediate signal transfer from the cell surface to the

4
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

Fig. 4. The chemical structures of indole alkaloids and indole derivatives that regulate autophagy.

5
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

intracellular. It plays a crucial role in the process of autophagy through endoplasmic reticulum (ER) stress [87]. Hu’s group isolated a series of
different mechanisms [68]. Three MAPK pathways involved in the indole notoamide-type alkaloids from a coral-associated fungus Asper­
regulation of cellular activities have been studied. They are the ERK1/2, gillus ochraceus LZDX-32–15. Notoamide G exerts potent cytotoxicity in
JNK, and p38/MAPK signaling pathways [69]. the HepG2 and Huh-7 cells via autophagy and apoptosis. Notoamide
ERK1/2 are activated by mitogens and transmit extracellular signals G-mediated autophagy is shown by the upregulation of Beclin-1 and
into the nucleus through cell-membrane receptors, thus mediating the LC3B, the key protein of autophagy [88]. Further investigation revealed
expression of the intracellular gene and participating in the regulation of that notoamide G promotes P38 and JNK phosphorylation. This suggests
cell proliferation, differentiation, apoptosis, autophagy, and other that notoamide G induces autophagy and apoptosis through P38/JNK
functions [70,71]. Numerous studies have shown that the activation of pathway, which indicates that notoamide G may serve as a potent lead
ERK1/2 can directly promote the upregulation of the autophagy marker for further development as an antitumor agent.
proteins LC3 and p62 to initiate autophagy [72]. Meanwhile, it can Abe and his coworkers reported that harmol, a β-carboline alkaloid,
cause the downregulation of lysosomal-associated membrane protein 1 exhibits significant antitumor activity in human NSCLC A549 cells. The
(LAMP1) and LAMP2, followed by the prevention of the binding of alkaloid promotes cell death by inducing autophagy rather than
autophagosomes to lysosomes, which inhibits the reduction of auto­ apoptosis, as shown by the increase in LC3II, a reliable molecular marker
phagosomes [73]. for autophagy detection. The induction is partly provoked by the ERK 1/
Another MAPK pathway protein is c-Jun N-terminal kinase (JNK). 2 signaling pathway [89]. Additionally, Ishimwe et al. [90] demon­
The JNK signaling pathway can induce autophagy through the Beclin-1- strated that brucine, a clinically-used small molecule in traditional
dependent or Beclin-1-independent pathway. On the one hand, the JNK Chinese medicine, exerts the effect of inhibition of autophagy. The
signaling pathway can phosphorylate Bcl-2 family proteins to promote inhibitory effect is achieved by disturbing autolysosomal degradation
the release of Beclin-1 from the Beclin-1/Bcl-2 complex and bind to and exerting the regulation of ERK1/2-mTOR-p70S6K signaling cascade
Vps34 to promote autophagy. On the other hand, JNK can promote [90]. The inhibition of autophagy plays a vital role in inducing immu­
autophagy by mediating the upregulation of Atg gene expression. The nogenic cell death, which improves its application in cancer treatment.
autophagy induced by the JNK pathway plays a protective role in pro­ Chaetoglobosin G is a fungal secondary metabolite and exhibits anti­
moting cancer-cell survival, which counters apoptosis [74–77]. cancer activity. Chen and his coworker showed that chaetoglobosin af­
The P38/MAPK signaling pathway is also extensively involved in fects the arrest of cell proliferation, and the inhibition is mediated by the
autophagy regulation. It comprises five subtypes, namely, p38α, p38β1, induction of autophagy and apoptosis, which is regulated by the
p38β2, p38γ, and p38δ. The p38/MAPK signaling pathway exerts a EGFR/MEK/ERK pathway. The result is the increased expression of P21
critical role in the regulation of cell-cycle arrest, apoptosis, autophagy, and LC3II, G2/M phase arrest, and decreased Bcl-2 protein [91]. Table 1.
growth inhibition, and differentiation [78]. The p38/MAPK signaling
pathway serves a dual role in autophagy regulation, namely positive 4. Targeting Beclin-1
regulator and negative regulator. Sometimes autophagy can be induced
by activating this signaling pathway [79,80]. Meanwhile, the Beclin-1 gene is the first mammalian autophagy gene to be found by
p38/MAPK signaling pathway has a negative role in autophagy regu­ yeast two-hybrid screening in 1998 by Beth Levine’s group. It is located
lation. Several studies have proven that autophagy can be induced by on chromosome 17q21 and is an ortholog of the autophagy gene Atg6/
inhibiting this pathway [81,82]. Thus, targeting ERK1/2, p38, and JNK Vps30 protein in yeast [92]. Beclin-1, a 60-kDa protein of 450 amino
MAPK to regulate autophagy is a strategy against cancer cells that is acids, possesses three identified functional domains: an evolutionarily
worth exploring. conserved domain (amino acids 244–337) [93], a central coiled-coil
An aspidosperma-type alkaloid, 11-methoxytabersonine (11-MT), domain (amino acids 144–269) [94], and a BH3 domain (amino acids
has been isolated from the roots of Tabernaemontana Bovina Lour. Ge 114–123) at the N-terminus [95]. As a major regulator, Beclin-1 regu­
et al. [83] demonstrated that 11-MT could induce protective autophagy lates autophagy by forming complexes with some other proteins. As an
in A549 and H157 cells to avoid 11-MT-induced necroptosis. Mechanism essential member of Class III PI3K complexes, Beclin-1 can produce two
research has suggested that the alkaloid regulates autophagy through types of complexes: PI3KC3-C1 and PI3KC3-C2. PI3KC3-C1 is crucial to
the AMPK/mTOR and JNK signaling pathways [83]. Shimizu and his autophagic vesicle enucleation at the start of autophagosome formation,
coworkers demonstrated that the JNK signaling pathways are crucial to comprising Beclin-1, Vps34, Vps15, and Atg14. PI3KC3-C2 is associated
autophagic cell death. In return, the activation of autophagy can trigger with autophagolysosomal maturation and contains Beclin-1, Vps34,
JNK [84]. Evodiamine is a naturally occurring indole alkaloid and is the Vps15, and UV Radiation Resistance Associated Gene Protein [96,97].
main biologically active component of the traditional Chinese medicine Beclin-1 was first identified as a Bcl-2 interacting partner, and Bcl-2
evodiae fructus. Liu et al. [85] revealed that evodiamine induces auto­ and Bcl-XL exert a critical role in autophagy regulation activity by
phagy and apoptosis in human glioblastoma cells [85]. The apoptosis combining with Beclin-1. They can inhibit autophagy by binding to the
induced by evodiamine is mediated by the calcium pathway, which is BH3 domain of Beclin-1 so that Beclin-1 cannot interact with other
shown by the decrease of cytosolic calcium elevation, apoptosis, and proteins to form the PI3KC3 complex [98]. Under normal conditions,
mitochondrial depolarization under calcium channels blockade. In Bcl-2 inhibits Beclin-1. However, under stress conditions, various signals
addition, blockade of calcium channels and inactivation of JNK inhibits can regulate the interaction between Bcl-2/Bcl-XL and Beclin-1. The
the autophagy-mediated by evodiamine, which suggests that evodi­ phosphorylation of Bcl-2/Bcl-XL and Beclin-1 is the major regulator to
amine mediates autophagy via a calcium-JNK signaling pathway. Yang regulate the combination. First, the phosphorylation of Bcl-2 may
et al. [86] synthesized a series of calothrixin A (CAA) analogs, one of regulate the Bcl-2-Beclin 1 complex. Under starvation conditions, JNK1
which is CAA45. Their investigation confirmed that CAA45 blocks can phosphorylate Bcl-2 at T69, S70, and S97, thereby contributing to
cell-cycle arrest, induces cell apoptosis and autophagy, and inhibits the the dissociation of Bcl-2-Beclin 1 and leading to the induction of auto­
cells migration. Notably, apoptosis and autophagy-mediated by CAA45 phagy [99]. Second, the phosphorylation of Beclin-1 plays a dual role in
are regulated by the Akt/JNK/p53 signaling pathway [86]. autophagy regulation. The overexpression of death-inducing kinase and
Isomahanine is an alkaloid containing an indole skeleton isolated serine/threonine Rho kinase 1 promotes the phosphorylation of Beclin-1
from Murraya koenigii and exhibits wide-ranging bioactivities. Utaipan at the threonine 119 site of the BH3 domain, thereby leading to the
et al. [87] found that isomahanine induces apoptosis and autophagy, dissociation from Bcl-2 and initiation of autophagy through Beclin-1
playing a prodeath role in the multidrug‑resistant oral squamous cell binding to the Vps34 complex [100,101]. Conversely, Beclin-1 can be
carcinoma (OSCC) cell line CLS-354/DX. These effects are associated phosphorylated at the T108 site of the BH3 domain by proapoptotic
with the activation of the P38/MAPK signaling pathway mediated by kinase mammalian Ste20-like kinase 1, but the interaction between

6
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

Table 1
Regulation of indole alkaloids and indole derivatives on autophagy.
Targets Compounds Cell lines Effect Dual Role References
onAutophagy ofAutophagy

PI3K/Akt/mTOR pathway Harmol U251MG cells Induces Pro-death [57]


Harmine B16 cells Induces Pro-death [59]
Harmine MGC-803 cells;SGC-7901 Induces Pro-death [60]
cells
Fascaplysin HL-60 cells Induces Pro-death [62]
Fascaplysin chloride MDAMB-231 cells Induces Pro-death [63]
Bisleuconothine A MCF7 cells;A549 cells Induces Pro-death [64]
Staurosporine Hepg2 cells Induces Pro-survival [65]
Indole-3-carbinol Zebrafish larvae Induces Pro-survival [66]
MAPK pathway JNK/MAPK pathways 11-methoxytabersonine A549 cells;H157 cells Induces Pro-survival [83]
JNK/MAPK pathways Evodiamine Glioblastoma cells Induces Pro-death [85]
JNK/MAPK pathways Calothrixin A 45 A549 cells Induces Unknown [86]
P38/MAPK pathways Isomahanine CLS-354/DX cells Induces Pro-death [87]
P38/MAPK pathways Notoamide G Hepg2 cells;Huh-7 cells Inhibits Unknown [88]
Erk1/2/MAPK pathways Harmol A549 Cells Induces Pro-death [89]
Erk1/2/MAPK pathways Brucine MDA-MB-231 cells;CT26 Inhibits Pro-death [90]
cells
Erk1/2/MAPK pathways Chaetoglobosin G A549cells Induces Pro-death [91]
Beclin-1 Evodiamine SGC-7901 Cells Induces Pro-death [104]
Vincristine Zebrafish larvae neuronal Inhibits Pro-death [105]
cells
Vincristine MDA-MB-231 Cells Induces Unknown [106]
Violacein Neck carcinoma cells Induces Pro-death [107]
IF203 HepG2 cells Inhibits Unknown [108]
ROS pathways Evodiamine Hela cells Inhibits Pro-survival [113]
Reduced scytonemin T-lymphoid Jurkat cell Induces Pro-death [114]
Indole alkaloid derivative B HT-29 cells Induces Pro-death [115]
(IADB)
Fascaplysin Vascular endothelia cells Induces Pro-survival [116]
P62 Reserpine HEK293 cell Inhibits Pro-death [121]
Racemocin B derivative C25 MDA-MB-231 cells Inhibits Pro-death [122]
Conophylline Fatty liver cells(in vivo) Induces Pro-survival [123]
Taberine D Hela cells Inhibits Pro-survival [124]
Indole-3-carbinol Ehrlich ascites carcinoma Inhibits Pro-death [125]
cells
Miscellaneous P53/AKT pathway Ellipticine A549 cells Induces Pro-death [126]
pathways NF-κB p65 pathway BMA155Cl HepG2 cells Induces Pro-death [128]
JAK2/STAT3 pathway Bufothionine HCC cells Induces Pro-death [129]
ER stress Aplysinopsin Derivative EE-84 K562 cells Induces Pro-death [130]
ER stress CTet MDA-MB-231 cells Induces Dual role [131]
Depending on the mTOR Corynoxine Neuronal cell lines Induces Pro-survival [132]
pathway

Beclin-1 and Bcl-2/Bcl-xl is promoted and autophagy is inhibited [102]. and neck cancer cells in vitro and in vivo by inducing autophagy and
Other regulators can also regulate the Bcl-2-Beclin-1 interaction, such as apoptosis. Violacein increased PARP-1 cleavage, Bax/Bcl-2 ratio, inhi­
a BH3-only protein/mimetic, which induces autophagy by disrupting bition of ERK1/2 phosphorylation, and light chain 3-II (LC3-II) expres­
the Bcl-2-Beclin-1 complex [103]. sion. Furthermore, violacein disturbs Bcl-2-Beclin-1 interaction by
The natural product evodiamine is an alkaloid isolated from the decreasing the expression of Bcl-2, thereby leading to upregulation of
traditional Chinese medicine evodiae fructus, which is the dried fruit of Beclin-1 expression, which serves as a key regulator in autophagy to
Evodia rutaecarpa Bentham. It exerts a deleterious effect on SGC-7901 regulation [107]. This is the first report demonstrating
human gastric adenocarcinoma cells. Evodiamine reportedly induces violacein-mediated autophagy in cancer cells, which provides a new
apoptosis, autophagy, and cell-cycle arrest at the G2/M phase. These mechanism for violacein as an anticancer molecule. Shang et al. [108]
effects lead to the downregulation of Bcl-2 and upregulation of Bax. demonstrated that IF203, a novel indole derivative, has an efficient
Moreover, Beclin-1 is associated with evodiamine-induced autophagy inhibitory effect on HepG2 cell growth, and this effect is caused by
[104]. Hu and his coworkers investigated the relationship between cell-cycle arrest, apoptosis, and autophagy induced by IF203. In addi­
autophagy and vincristine neurotoxicity. They found that vincristine tion, autophagy activation is triggered by the positive expression of the
inhibits autophagy but induces apoptosis in the dopaminergic neurons autophagy-related proteins Atg5, Atg12, ULK1, Beclin-1, and LC3II
of zebrafish, leading to neurotoxicity. Here, Beclin-1 plays a dominant [108].
role in regulating autophagy, suggesting that Beclin-1 may be a potential
molecular target to reduce vincristine neurotoxicity [105]. Furthermore, 5. Targeting the ROS signaling pathway
Sun et al. [106] demonstrated that vincristine blocks the combination of
sirt 2 and heat shock protein (Hsp)70, which leads to Hsp70 acetylation Reactive oxygen species (ROS) is a general term for oxygen-
at K126. The acetylation of Hsp 70 enhances the combination of Hsp 70 containing free radicals and peroxides that easily form free radicals
and Bcl-2, thereby promoting the upregulation of Beclin-1 and playing a related to oxygen metabolism in the organism. As a product of normal
pivotal role in autophagy [106]. Masuelli et al. [107] demonstrated that redox reactions in the body, ROS is involved in the regulation of steril­
the indole-derived violacein, a purple-colored natural pigment, exerts ization, detoxification, and various metabolic pathways [109]. ROS
remarkable activities to inhibit a panel of head and neck carcinoma cell include oxygen anions, free radicals, and hydrogen peroxide [110] and
lines. This study revealed that violacein could inhibit the growth of head originates from two major pathways: mitochondria and nicotinamide

7
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

adenine dinucleotide phosphate (NADPH) oxidase [111]. Reserpine, an indole alkaloid isolated initially from the snakeroot of
ROS, especially mitochondria-derived ROS, can reportedly regulate the rauchia plant, is used to treat hypertension and schizophrenia. Lee
autophagy [112]. The molecular mechanism of ROS regulation of et al. [121] demonstrated that reserpine blocks autophagic flux, leading
autophagy primarily depends on various molecular signal pathways, to cell death in GFP-LC3 cells and neuronal cells through the upregu­
such as ROS-FOXO3-LC3/BNIP3-autophagy, ROS-NRF2-P62-autophagy, lation of LC3II and p62/SQSTM1 expression. In neuronal cells, reserpine
ROS-HIF1-BNIP3/NIX-autophagy, and ROS-TIGAR-autophagy. The induces the accumulation of α-synuclein protein, which is crucial to PD
accumulation of ROS leads to the oxidative-stress response and activates pathogenesis by inhibiting autophagy flux. Interestingly, treating
HIF-1, p53, FOXO3, and NRF2, respectively. These proteins induce the neuronal cells with the combination of reserpine and rapamycin can
transcription of BNIP3 and NIX, TIGAR, LC3, and BNIP3 and p62, enhance the effect of reserpine, leading to reduced cell viability [121].
respectively. The corresponding protein regulates autophagy through Xiao and his coworkers synthesized a series of novel derivatives of
various autophagy pathways, such as MAPKs, mTOR, and AMPK/Akt. racemocin B, an indole natural product isolated from the green alga
Evodiamine is an alkaloid isolated from the dried, unripe fruit of Caulerpa racemose. C25 possesses potent anticancer activity, effectively
Evodia rutaecarpa (Juss.) Benth. It shows remarkable activities inhib­ inducing cell-cycle arrest, apoptosis, and cell death in the triple-negative
iting human cervix carcinoma HeLa cells. It induces apoptosis, which breast cancer cell line MDA-MB-231. Inhibition of autophagy plays a
plays a critical role in cell death and is dependent on the ROS pathway. role in these effects by inducing the expression of LC3II and
Notably, moderate ROS/NO leads to autophagy, protecting against cell p62/SQSTM1, the marker of blocking autophagic flux [122]. Con­
death. However, massive ROS/NO plays a negative role in autophagy, ophylline is a naturally occurring indole alkaloid present in the leaves of
which promotes cell death [113]. Reduced scytonemin (R-scy), isolated the tropical plant Tabernaemontana divaricate. Conophylline can
from N. commune Vaucher, reportedly possesses a higher inhibitory reportedly alleviate hepatic steatosis by promoting the expression of
activity in human T-lymphoid Jurkat cells. The cytotoxicity of R-scy peroxisome proliferator-activated receptor alpha (PPARA), carnitine
depends on the autophagic death induced by the natural compound. palmityl transferase (CPT)1, and CPT2-related β-oxidation. It can also
ROS pathway plays pivotal roles in cell death mediated by R-scy, which induce autophagy by regulating Atg7 and p62, which play crucial roles
is involved in the induction of mitochondrial dysfunction [114]. in hepatic lipotoxicity, thereby providing a novel molecular mechanism
Bi and his coworkers synthesized a new indole derivative B (IADB) to prevent hepatic steatosis in non-alcoholic fatty liver disease (NAFLD)
which shows potent cytotoxicity to tumor cells and is nontoxic to cardiac [123].
cells [115]. It selectively induces autophagic cell death mediated by ROS Zhang et al. [124] obtained several bisindole alkaloids from Taber­
overproduction in cancer cells in colon cancer HT29 cells due to dif­ naemontana corymbose. They also found that taberine D causes
ferences in redox status between normal and tumor cells. In normal cells, dysfunction in lysosomes by attenuating lysosomal acidification, which
IADB treatment in combination with chemotherapy can mitigate is involved in the block of autophagic flux in HeLa cells. The blocking
5–Fluorouracil-Induced Cardiotoxicity. The study about IADB may process is shown by the increase in LC3II and p62/SQSTM1 [124]. I3C is
support novel therapeutic targets to improve chemotherapeutic efficacy an active natural product of cruciferous vegetables, which is known as
with decreased off-target effects. Meng and his partner demonstrated an antitumor substance. Donia and his partner reported that the coad­
that fascaplysin, a red pigment isolated from a Fijian marine sponge ministration of I3C with hydroxychloroquine exerts a deleterious effect
(Fascaplysinopsis sp.), provokes autophagy in vascular endothelial cells on ehrlich ascites carcinoma cells by regulating apoptosis and auto­
by activating the ROS pathway. ROS pathway-mediated autophagy phagy. The process is mediated by significant upregulation in LC3B and
plays a protective role in vascular endothelia cells to inhibit apoptosis downregulation in p62 gene expression [125].
and its anti-angiogenesis activity, providing a novel anticancer appli­
cation of fascaplysin [116]. 7. Targeting miscellaneous pathways

6. Targeting P62/SQSTM1 Apart from the pathways described above, to exert antitumor ac­
tivity, indole alkaloids and their derivatives can also regulate autophagy
P62/SQSTM1 is a multifunctional protein and exerts multiple effects through other pathways, such as the P53/Akt and JAK2/STAT3 path­
on cell-signaling transduction, cell proliferation, apoptosis, and tumor­ ways, as well as ER stress. Fang and his partners demonstrated that
igenesis [117]. As the main component of many protein complexes in ellipticine, a famous topoisomerase II inhibitor, has higher inhibitory
cells, P62 contains multiple distinct domains, regulating different activity in human NSCLC cells. It can induce autophagy and apoptosis
signaling pathways by interacting with other proteins [118], such as the and arrest the cell cycle at the G2/M phase, associated with ellipticine-
Keap1-Nrf2, mTOR, and NF-κB pathways. induced cytotoxicity. The autophagy caused by ellipticine is also
P62 is known to play an important role in autophagy through its mediated by the corporation of p53 and Akt [126]. Afterward, the same
series of distinct domains, including ubiquitin-associated (UBA), LC3- group further proved that the presence of P53 facilitates the nucleus
interacting region (LIR), and N-terminal phox-BEM1 (PB1) domains, translocation of phosphorylated Akt and the activation of the autophagy
which contribute to the binding of p62 to ubiquitin, LC3, and many pathway, thereby leading to apoptotic cell death [127]. These studies
other proteins [119]. Ser407 in the UBA domain of p62 is phosphory­ provide a new molecular mechanism for ellipticine as an effective
lated by autophagy-associated protein 1. Subsequently, Ser403 in the antitumor therapy agent. BMA155 is a derivative of bisindolylmalei­
UBA domain is phosphorylated by Casein kinase 2 or Tank-binding ki­ mide alkaloid synthesized from the indole skeleton, which exhibits su­
nase 1, leading to p62 ubiquitination and thus promoting autophagy perior inhibitory activity in HepG-2 cells. Sun et al. [128] proved that
degradation [118]. The PB1 domain of P62 can promote the packaging BMA-155Cl, which is easily obtained from BMA-155, has a remarkable
of ubiquitinated substrates through self-oligomerization and subse­ antitumor effect by inducing apoptosis and autophagy in human hep­
quently transport the packaged substrates to the autophagy pathway to atocarcinoma HepG-2 cells. In the process, BMA-155Cl induces auto­
participate in autophagosome formation. P62 interacts with LC3 phagy by activating the NF-κB p65 pathway, promoting the subsequent
through its LIR domain to form a complex, which is degraded together in apoptosis and serving as a protective method [128].
autophagy lysozyme as an autophagy-specific substrate [120]. P62 is Bufothionine is known as the main active ingredient of cinobufacini,
degraded by autophagy and in turn regulates the autophagy process. which is isolated from the skins and parotid venom glands of the toad
During normal autophagy, p62 protein is continuously degraded in the Bufo gargarizans Cantor and Bufo melanostictus Schneider. Kong et al.
cytoplasm. When autophagy activity is weakened, and autophagy [129] reported that bufothionine could inhibit cell proliferation and
function is defective, P62 protein continuously accumulates in the promote apoptosis and autophagy in SMMC7721 cells. The activation of
cytoplasm, so P62 is a marker protein that reflects autophagy activity. autophagy is suggested by the positive expression of autophagy-related

8
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

proteins, such as Atg5, Atg7, Beclin-1, and LC3II. The inhibition of the Acknowledgment
JAK2/STAT3 pathway plays an important role in bufothionine-induced
autophagy [129]. Song et al. [130] demonstrated that the aplysinopsin We are grateful for financial support from National Natural Science
derivative EE-84 could induce cellular morphologic change and Foundation of China (82073998, 22107015), Sichuan Science and
cell-stress responses, which trigger autophagy and ER stress in chronic Technology Program (2022JDRC0045, 2022JDRC0125, 2019YJ0321),
myeloid leukemia cells. Moreover, EE84-induced autophagy is related to the China Postdoctoral Science Foundation and Xinglin Scholar
the PERK-eIF2a-ATF4-CHOP UPR pathway, which is provoked by Research Promotion Project of Chengdu University of TCM.
EE84-induced ER stress. These results show that EE84 is expected to be a
potential drug agent for leukemia treatment due to its excellent prop­ Conflict of interest statement
erties of antileukemia [130]. I3C and its metabolic products are
considered as the main anticancer active ingredient from cruciferous The authors declare that there are no conflicts of interest.
vegetables. Galluzzi’s group showed that the autophagy induced by CTet
is activated by ER stress in MDA-MB-231 cells. Meanwhile, References
CTet-mediated autophagy plays a protective role at early time points,
resulting in autophagic cell death with time [131]. Another group re­ [1] H. Sung, J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal,
ported that conophylline, a vinca alkaloid, can also induce autophagy, F. Bray, Global cancer statistics 2020: globocan estimates of incidence and
mortality worldwide for 36 cancers in 185 countries, CA Cancer J. Clin. 71 (2021)
suppress protein aggregation, and protect cells from cell death. Notably, 209–249, https://doi.org/10.3322/caac.21660.
conophylline-induced autophagy does not depend on the mTOR [2] T. Minko, L. Rodriguez-Rodriguez, V. Pozharov, Nanotechnology approaches for
pathway; instead, it is induced independently of the mTOR pathway personalized treatment of multidrug resistant cancers, Adv. Drug Deliv. Rev. 65
(2013) 1880–1895, https://doi.org/10.1016/j.addr.2013.09.017.
[132]. [3] N. Wang, Y. Feng, N. Wang, Y. Feng, Elaborating the role of natural products-
induced autophagy in cancer treatment: achievements and artifacts in the state of
8. Conclusion and perspective the art, BioMed. Res. Int. 2015 (2015), 934207, https://doi.org/10.1155/2015/
934207.
[4] T. Xie, S. Song, S. Li, L. Ouyang, L. Xia, J. Huang, Review of natural product
As an evolutionarily conserved mechanism, Autophagy is rather databases, Cell Prolif. 48 (2015) 398–404, https://doi.org/10.1111/cpr.12190.
complex but essential under basal conditions and settings of diseases. [5] X. Zhang, L.X. Chen, L. Ouyang, Y. Cheng, B. Liu, Plant natural compounds:
targeting pathways of autophagy as anti-cancer therapeutic agents, Cell Prolif. 45
Over the past few years, increasing indole-containing alkaloids are
(2012) 466–476, https://doi.org/10.1111/j.1365-2184.2012.00833.x.
emerging with autophagy-regulating capacity in experimental anti­ [6] W. Tan, J. Lu, M. Huang, Y. Li, Anti-cancer natural products isolated from chinese
tumor therapies. Actually, a large body of evidence has indicated that medicinal herbs, Chin. Med 27 (2011) 1–15, https://doi.org/10.1186/1749-
indole alkaloid-mediated autophagy plays dual roles in cancer. In this 8546-6-27.
[7] C.M. Giuliani, C.R. Dass, Autophagy and cancer: taking the ‘toxic’ out of
review, we summarize the pharmacological action of indole alkaloids cytotoxics, J. Pharm. Pharm. 65 (2013) 777–789, https://doi.org/10.1111/
and derivatives and their regulation mechanism of autophagy based on jphp.12034.
previous findings. As summarized in our article, indole alkaloids regu­ [8] J.C. Tanaka, C.C. Silva, A.J. Oliveira, C.V. Nakamura, B.P. Dias Filho,
Antibacterial activity of indole alkaloids from aspidosperma ramiflorum, Braz. J.
late autophagy by modulating the activity of various signaling path­ Med. Biol. Res. 39 (2006) 387–391, https://doi.org/10.1590/s0100-
ways, including those involving PI3K/Akt/mTOR, MAPK, Beclin-1, and 879x2006000300009.
ROS. As discussed above, some indole alkaloids activate autophagic cell [9] R.B. Williams, J.F. Hu, K.M. Olson, V.L. Norman, M.G. Goering, M. O’Neil-
Johnson, G.R. Eldridge, C.M. Starks, Antibiotic indole sesquiterpene alkaloid
death, whereas some others inhibit it. Furthermore, as discussed above, from greenwayodendron suaveolens with a new natural product framework,
the same indole alkaloid can activate different signaling pathways to J. Nat. Prod. 73 (2010) 1008–1011, https://doi.org/10.1021/np1001225.
regulate autophagy. [10] S.Y. Long, C.L. Li, J. Hu, Q.J. Zhao, D. Chen, Indole alkaloids from the aerial parts
of Kopsia fruticosa and their cytotoxic, antimicrobial and antifungal activities,
All studies described above fails to explore further the causative role
Fitoterapia 129 (2018) 145–149, https://doi.org/10.1016/j.fitote.2018.06.017.
of autophagy in the anticancer effect of indole alkaloids. This role re­ [11] M.L. Yang, J. Chen, M. Sun, D.B. Zhang, K. Gao, Antifungal indole alkaloids from
quires determining whether autophagy is a major independent cell winchia calophylla, Planta Med 82 (2016) 712–716, https://doi.org/10.1055/s-
0042-102459.
killer, an early phase of cell death occurring prior to apoptotic cell death,
[12] L.P. Robertson, L. Lucantoni, V.M. Avery, A.R. Carroll, Antiplasmodial bis-indole
or a passive bystander with an effect that occurs concomitantly with alkaloids from the bark of flindersia pimenteliana, Planta Med 86 (2020) 19–25,
apoptotic cell death. Thus, an increased understanding of autophagy https://doi.org/10.1055/a-1028-7786.
associated with indole alkaloids with cancer therapy is urgently [13] D.B. Tlhapi, I.D.I. Ramaite, T. Van Ree, C.P. Anokwuru, T.S. Orazio, H.C. Hoppe,
Isolation, chemical profile and antimalarial activities of bioactive compounds
required. At present, most mechanistic studies have focused on in vitro from rauvolfia caffra sond, Molecules 24 (2018) 39–49, https://doi.org/10.3390/
cellular models. Conversely, the establishment of appropriate experi­ molecules24010039.
mental models to evaluate the role of autophagy in vivo also has [14] T.Z. Xie, Y.L. Zhao, J.J. He, L.X. Zhao, X. Wei, Y.P. Liu, X.D. Luo, Monoterpenoid
indole alkaloids from the stems of kopsia officinalis, Fitoterapia 143 (2020),
fundamental significance in evaluating the therapeutic effects and safety 104547, https://doi.org/10.1016/j.fitote.2020.104547.
as new anticancer agents for cancer therapy. In summary, we hope that [15] L. Fang, S.M. Tian, J. Zhou, Y.L. Lin, Z.W. Wang, X. Wang, Melaxillines A and B,
more indole-contain compounds or naturally indole alkaloids can be monoterpenoid indole alkaloids from melodinus axillaris, Fitoterapia 115 (2016)
173–176, https://doi.org/10.1016/j.fitote.2016.10.012.
found and synthesized. The autophagy-regulation mechanism and [16] Y.P. Liu, Q.L. Liu, X.L. Zhang, H.Y. Niu, C.Y. Guan, F.K. Sun, W. Xu, Y.H. Fu,
structure-activity relationship of these compounds can be deeply Bioactive monoterpene indole alkaloids from nauclea officinalis, Bioorg. Chem.
explored to shed light on the development of strategies for cancer 83 (2019) 1–5, https://doi.org/10.1016/j.bioorg.2018.10.013.
[17] D. Zhang, L. Zhao, L. Wang, X. Fang, J. Zhao, X. Wang, L. Li, H. Liu, Y. Wei,
treatment.
X. You, S. Cen, L. Yu, Griseofulvin derivative and indole alkaloids from
penicillium griseofulvum CPCC 400528, J. Nat. Prod. 80 (2017) 371–376,
CRediT authorship contribution statement https://doi.org/10.1021/acs.jnatprod.6b00829.
[18] G. Philippe, P. De Mol, M. Zèches-Hanrot, J.M. Nuzillard, M.H. Tits, L. Angenot,
M. Frédérich, Indolomonoterpenic alkaloids from strychnos Icaja roots,
Bo Han and Qian Zhao contributed to the conception and design of Phytochemistry 62 (2003) 623–629, https://doi.org/10.1016/s0031-9422(02)
the study. Meng-Lan Luo and Wei Huang organized the database, per­ 00612-x.
formed the statistical analysis, and wrote the first draft of the manu­ [19] R. Verpoorte, M. Frédérich, C. Delaude, L. Angenot, G. Dive, P. Thépenier, M.-
J. Jacquier, M. Zèches-Hanrot, C. Lavaud, J.-M. Nuzillard, Moandaensine, a
script. Hong-Ping Zhu and Cheng Peng contributed to the manuscript dimeric indole alkaloid from strychnos moandaensis (Loganiaceae), Phytochem.
revision. All authors read and approved the submitted version. Lett. 3 (2010) 100–103, https://doi.org/10.1016/j.phytol.2010.02.005.
[20] C. Toso, C. Lindley, Vinorelbine: a novel vinca alkaloid, Am. J. Health Syst.
Pharm. 52 (1995) 1287–1304, https://doi.org/10.1093/ajhp/52.12.1287.
[21] M.E. van de Velde, G.L. Kaspers, F.C.H. Abbink, A.J. Wilhelm, J.C.F. Ket, M.
H. van den Berg, Vincristine-induced peripheral neuropathy in children with

9
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

cancer: a systematic review, Crit. Rev. Oncol. 114 (2017) 114–130, https://doi. [42] N. Mizushima, M. Komatsu, Autophagy: renovation of cells and tissues, Cell 147
org/10.1016/j.critrevonc.2017.04.004. (2011) 728–741, https://doi.org/10.1016/j.cell.2011.10.026.
[22] A. Capasso, Vinorelbine in cancer therapy, Curr. Drug Targets 13 (2012) [43] Y. Ohsumi, Molecular dissection of autophagy: two ubiquitin-like systems, Nat.
1065–1071, https://doi.org/10.2174/138945012802009017. Rev. Mol. Cell Biol. 2 (2001) 211–216, https://doi.org/10.1038/35056522.
[23] W. Abida, D. Campbell, A. Patnaik, J.D. Shapiro, B. Sautois, N.J. Vogelzang, E. [44] L. Yu, Y. Chen, S.A. Tooze, Autophagy pathway: cellular and molecular
G. Voog, A.H. Bryce, R. McDermott, F. Ricci, J. Rowe, J. Zhang, J.M. Piulats, mechanisms, Autophagy 14 (2018) 207–215, https://doi.org/10.1080/
K. Fizazi, A.S. Merseburger, C.S. Higano, L.E. Krieger, C.J. Ryan, F.Y. Feng, A. 15548627.2017.1378838.
D. Simmons, A. Loehr, D. Despain, M. Dowson, F. Green, S.P. Watkins, [45] S. Chen, S.K. Rehman, W. Zhang, A. Wen, L. Yao, J. Zhang, Autophagy is a
T. Golsorkhi, S. Chowdhury, Non-BRCA DNA damage repair gene alterations and therapeutic target in anticancer drug resistance, Biochim. Biophys. Acta 2010
response to the PARP inhibitor rucaparib in metastatic castration-resistant (1806) 220–229, https://doi.org/10.1016/j.bbcan.2010.07.003.
prostate cancer: analysis from the phase ІІ TRITON2 study, Clin. Cancer Res. 26 [46] C.H. Jung, S.H. Ro, J. Cao, N.M. Otto, D.H. Kim, mTOR regulation of autophagy,
(2020) 2487–2496, https://doi.org/10.1158/1078-0432.Ccr-20-0394. FEBS Lett. 584 (2010) 1287–1295, https://doi.org/10.1016/j.
[24] D.R. Camidge, R. Dziadziuszko, S. Peters, T. Mok, J. Noe, M. Nowicka, S. febslet.2010.01.017.
M. Gadgeel, P. Cheema, N. Pavlakis, F. de Marinis, B.C. Cho, L. Zhang, D. Moro- [47] B. Karakas, K.E. Bachman, B.H. Park, Mutation of the PIK3CA oncogene in human
Sibilot, T. Liu, W. Bordogna, B. Balas, B. Müller, A.T. Shaw, Updated efficacy and cancers, Br. J. Cancer 94 (2006) 455–459, https://doi.org/10.1038/sj.
safety data and impact of the EML4-ALK fusion variant on the efficacy of alectinib bjc.6602970.
in untreated ALK-positive advanced non-small cell lung cancer in the global phase [48] Y. Samuels, Z. Wang, A. Bardelli, N. Silliman, J. Ptak, S. Szabo, H. Yan, A. Gazdar,
ІІІ ALEX study, J. Thorac. Oncol. 14 (2019) 1233–1243, https://doi.org/ S.M. Powell, G.J. Riggins, J.K. Willson, S. Markowitz, K.W. Kinzler, B. Vogelstein,
10.1016/j.jtho.2019.03.007. V.E. Velculescu, High frequency of mutations of the PIK3CA gene in human
[25] G.R. Oxnard, Y. Hu, K.F. Mileham, H. Husain, D.B. Costa, P. Tracy, N. Feeney, L. cancers, Science 304 (2004) 554, https://doi.org/10.1126/science.1096502.
M. Sholl, S.E. Dahlberg, A.J. Redig, D.J. Kwiatkowski, M.S. Rabin, C.P. Paweletz, [49] P. Liu, H. Cheng, T.M. Roberts, J.J. Zhao, Targeting the phosphoinositide 3-Ki­
K.S. Thress, P.A. Jänne, Assessment of resistance mechanisms and clinical nase pathway in cancer, Nat. Rev. Drug Disco 8 (2009) 627–644, https://doi.org/
implications in patients with EGFR T790M-positive lung cancer and acquired 10.1038/nrd2926.
resistance to osimertinib, JAMA Oncol. 4 (2018) 1527–1534, https://doi.org/ [50] D.A. Guertin, D.M. Sabatini, The pharmacology of mtor inhibition, Sci. Signal. 2
10.1001/jamaoncol.2018.2969. (2009) 24, https://doi.org/10.1126/scisignal.267pe24.
[26] J. Lu, H. Zhong, T. Chu, X. Zhang, R. Li, J. Sun, R. Zhong, Y. Yang, M.S. Alam, [51] B.A. Hemmings, D.F. Restuccia, PI3K-PKB/Akt pathway, Cold Spring Harb.
Y. Lou, J. Xu, Y. Zhang, J. Wu, X. Li, X. Zhao, K. Li, L. Lu, B. Han, Role of Perspect. Biol. 4 (2012) a011189, https://doi.org/10.1101/cshperspect.a011189.
anlotinib-induced CCL2 decrease in anti-angiogenesis and response prediction for [52] E. Cuyas, B. Corominas-Faja, J. Joven, J.A. Menendez, Cell cycle regulation by the
nonsmall cell lung cancer therapy, Eur. Respir. J. 53 (2019) 1801562, https://doi. nutrient-sensing mammalian target of rapamycin (mTOR) pathway, Methods Mol.
org/10.1183/13993003.01562-2018. Biol. 1170 (2014) 113–144, https://doi.org/10.1007/978-1-4939-0888-2_7.
[27] Z. Jia, Z. Zhang, Q. Tian, H. Wu, Y. Xie, A. Li, H. Zhang, Z. Yang, X. Zhang, [53] T. Noda, Y. Ohsumi, TOR a phosphatidylinositol kinase homologue, controls
Integration of transcriptomics and metabolomics reveals anlotinib-induced autophagy in yeast, J. Biol. Chem. 273 (1998) 3963–3966, https://doi.org/
cytotoxicity in colon cancer cells, Gene 786 (2021), 145625, https://doi.org/ 10.1074/jbc.273.7.3963.
10.1016/j.gene.2021.145625. [54] I. Dikic, Z. Elazar, Mechanism and medical implications of mammalian
[28] B. Wang, F. Peng, W. Huang, J. Zhou, N. Zhang, J. Sheng, P. Haruehanroengra, autophagy, Nat. Rev. Mol. Cell Biol. 19 (2018) 349–364, https://doi.org/
G. He, B. Han, Rational drug design, synthesis, and biological evaluation of novel 10.1038/s41580-018-0003-4.
chiral tetrahydronaphthalene-fused spirooxindole as MDM2-CDK4 dual inhibitor [55] B. Levine, G. Kroemer, Biological functions of autophagy genes: a disease
against glioblastoma, Acta Pharm. Sin. B. 10 (2020) 1492–1510, https://doi.org/ perspective, Cell 176 (2019) 11–42, https://doi.org/10.1016/j.cell.2018.09.048.
10.1016/j.apsb.2019.12.013. [56] F. Cecconi, B. Levine, The role of autophagy in mammalian development: cell
[29] Q. Zhao, C. Peng, H. Huang, S.J. Liu, Y.J. Zhong, W. Huang, G. He, B. Han, makeover rather than cell death, Dev. Cell. 15 (2008) 344–357, https://doi.org/
Asymmetric synthesis of tetrahydroisoquinoline-fused spirooxindoles as Ras-GTP 10.1016/j.devcel.2008.08.012.
inhibitors that inhibit colon adenocarcinoma cell proliferation and invasion, [57] A. Abe, H. Kokuba, Harmol induces autophagy and subsequent apoptosis in
Chem. Commun. 54 (2018) 8359–8362, https://doi.org/10.1039/c8cc04732d. U251MG human glioma cells through the downregulation of survivin, Oncol.
[30] M.C. Yang, C. Peng, H. Huang, L. Yang, X.H. He, W. Huang, H.L. Cui, G. He, Rep. 29 (2013) 1333–1342, https://doi.org/10.3892/or.2013.2242.
B. Han, Organocatalytic asymmetric synthesis of spiro-oxindole piperidine [58] Z.Q. Ma, H. Hu, T.T. He, H. Guo, M.Y. Zhang, M.W. Chen, Y.T. Wang, An
derivatives that reduce cancer cell proliferation by inhibiting MDM2-p53 assessment of traditional uighur medicine in current xinjiang region (China),
interaction, Org. Lett. 19 (2017) 6752–6755, https://doi.org/10.1021/acs. J. Tradit. Complement. Med. 11 (2014) 301–314, https://doi.org/10.4314/
orglett.7b03516. ajtcam.v11i2.13.
[31] X. Tang, N. Zhang, G. He, C.H. Li, W. Huang, X.Y. Wang, G. Zhan, B. Han, [59] N. Zou, Y. Wei, F. Li, Y. Yang, X. Cheng, C. Wang, The inhibitory effects of
Unconventional [2+3] cyclization involving [1,4]-sulfonyl transfer to construct compound Muniziqi granule against B16 cells and harmine induced autophagy
polysubstituted fluorazones as inhibitors of indoleamine 2,3-dioxygenase 1, Org. and apoptosis by inhibiting Akt/mTOR pathway, BMC Complement Altern. Med
Lett. 22 (2020) 7909–7914, https://doi.org/10.1021/acs.orglett.0c02836. 17 (2017) 517, https://doi.org/10.1186/s12906-017-2017-4.
[32] R. Zhou, Q. Wu, M. Guo, W. Huang, X. He, L. Yang, F. Peng, G. He, B. Han, [60] C. Li, Y. Wang, C. Wang, X. Yi, M. Li, X. He, Anticancer activities of harmine by
Organocatalytic cascade reaction for the asymmetric synthesis of novel chroman- inducing a pro-death autophagy and apoptosis in human gastric cancer cells,
fused spirooxindoles that potently inhibit cancer cell proliferation, Chem. Phytomedicine 28 (2017) 10–18, https://doi.org/10.1016/j.
Commun. 51 (2015) 13113–13116, https://doi.org/10.1039/c5cc04968g. phymed.2017.02.008.
[33] B. Han, X.H. He, Y.Q. Liu, G. He, C. Peng, J.L. Li, Asymmetric organocatalysis: an [61] S.B. Bharate, S. Manda, N. Mupparapu, N. Battini, R.A. Vishwakarma, Chemistry
enabling technology for medicinal chemistry, Chem. Soc. Rev. 50 (2021) and biology of fascaplysin, a potent marine-derived CDK-4 inhibitor, Mini Rev.
1522–1586, https://doi.org/10.1039/d0cs00196a. Med. Chem. 12 (2012) 650–664, https://doi.org/10.2174/
[34] S.J. Liu, Q. Zhao, C. Peng, Q. Mao, F. Wu, F.H. Zhang, Q.S. Feng, G. He, B. Han, 138955712800626719.
Design, synthesis, and biological evaluation of nitroisoxazole-containing spiro [62] S. Kumar, S.K. Guru, A.S. Pathania, S. Manda, A. Kumar, S.B. Bharate, R.
[pyrrolidin-oxindole] derivatives as novel glutathione peroxidase 4/mouse A. Vishwakarma, F. Malik, S. Bhushan, Fascaplysin induces caspase mediated
double minute 2 dual inhibitors that inhibit breast adenocarcinoma cell crosstalk between apoptosis and autophagy through the inhibition of PI3K/Akt/
proliferation, Eur. J. Med. Chem. 217 (2021), 113359, https://doi.org/10.1016/j. mTOR signaling cascade in human leukemia HL-60 cells, J. Cell. Biochem. 116
ejmech.2021.113359. (2015) 985–997, https://doi.org/10.1002/jcb.25053.
[35] Z. Yang, D.J. Klionsky, Eaten alive: a history of macroautophagy, Nat. Cell Biol. [63] S. Sharma, S.K. Guru, S. Manda, A. Kumar, M.J. Mintoo, V.D. Prasad, P.
12 (2010) 814–822, https://doi.org/10.1038/ncb0910-814. R. Sharma, D.M. Mondhe, S.B. Bharate, S. Bhushan, A marine sponge alkaloid
[36] R. Singh, A.M. Cuervo, Autophagy in the cellular energetic balance, Cell Metab. derivative 4-chloro fascaplysin inhibits tumor growth and vegf mediated
13 (2011) 495–504, https://doi.org/10.1016/j.cmet.2011.04.004. angiogenesis by disrupting PI3K/Akt/mTOR signaling cascade, Chem. Biol.
[37] A.E. Majeski, J.F. Dice, Mechanisms of chaperone-mediated autophagy, Int. J. Interact. 275 (2017) 47–60, https://doi.org/10.1016/j.cbi.2017.07.017.
Biochem. Cell B. 36 (2004) 2435–2444, https://doi.org/10.1016/j. [64] C.P. Wong, A. Seki, K. Horiguchi, T. Shoji, T. Arai, A.E. Nugroho, Y. Hirasawa,
biocel.2004.02.013. F. Sato, T. Kaneda, H. Morita, Bisleuconothine A induces autophagosome
[38] L. Marzella, J. Ahlberg, H. Glaumann, Autophagy, heterophagy, microautophagy formation by interfering with Akt-mTOR signaling pathway, J. Nat. Prod. 78
and crinophagy as the means for intracellular degradation, Virchows Arch. 36 (2015) 1656–1662, https://doi.org/10.1021/acs.jnatprod.5b00258.
(1981) 219–234, https://doi.org/10.1007/bf02912068. [65] Y. Ding, B. Wang, X. Chen, Y. Zhou, J. Ge, Staurosporine suppresses survival of
[39] D.J. Klionsky, S.D. Emr, Autophagy as a regulated pathway of cellular HepG2 cancer cells through Omi/HtrA2-mediated inhibition of PI3K/Akt
degradation, Science 290 (2000) 1717–1721, https://doi.org/10.1126/ signaling pathway, Tumor Biol. 39 (2017), https://doi.org/10.1177/
science.290.5497.1717. 1010428317694317.
[40] E.Y. Chan, A. Longatti, N.C. McKnight, S.A. Tooze, Kinase-inactivated ULK [66] Y. Jiang, G. Yang, Q. Liao, Y. Zou, Y. Du, J. Huang, Indole-3-carbinol inhibits lipid
proteins inhibit autophagy via their conserved C-terminal domains using an deposition and promotes autophagy in hyperlipidemia zebrafish larvae, Environ.
Atg13-independent mechanism, Mol. Cell Biol. 29 (2009) 157–171, https://doi. Toxicol. Pharmacol. 70 (2019), 103205, https://doi.org/10.1016/j.
org/10.1128/mcb.01082-08. etap.2019.103205.
[41] E.Y. Chan, mTORC1 phosphorylates the ULK1-mAtg13-FIP200 autophagy [67] G.P. Sreekanth, P.T. Yenchitsomanus, T. Limjindaporn, Role of mitogen-activated
regulatory complex, Sci. Signal. 2 (2009) 51, https://doi.org/10.1126/ protein kinase signaling in the pathogenesis of dengue virus infection, Cell.
scisignal.284pe51. Signal. 48 (2018) 64–68, https://doi.org/10.1016/j.cellsig.2018.05.002.

10
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

[68] T.A. Khavari, J. Rinn, Ras/Erk MAPK signaling in epidermal homeostasis and [92] X.H. Liang, L.K. Kleeman, H.H. Jiang, G. Gordon, J.E. Goldman, G. Berry,
neoplasia, Cell Cycle 6 (2007) 2928–2931, https://doi.org/10.4161/ B. Herman, B. Levine, Protection against fatal sindbis virus encephalitis by Beclin,
cc.6.23.4998. a novel Bcl-2-interacting protein, J. Virol. 72 (1998) 8586–8596, https://doi.org/
[69] I. Gkouveris, N.G. Nikitakis, Role of JNK signaling in oral cancer: a mini review, 10.1128/jvi.72.11.8586-8596.1998.
Tumor Biol. 39 (2017), https://doi.org/10.1177/1010428317711659. [93] N.N. Noda, T. Kobayashi, W. Adachi, Y. Fujioka, Y. Ohsumi, F. Inagaki, Structure
[70] S. Meloche, J. Pouyssegur, The ERK 1/2 mitogen-activated protein kinase of the novel c-terminal domain of vacuolar protein sorting 30/autophagy-related
pathway as a master regulator of the G1- to S-phase transition, Oncogene 26 protein 6 and its specific role in autophagy, J. Biol. Chem. 287 (2012)
(2007) 3227–3239, https://doi.org/10.1038/sj.onc.1210414. 16256–16266, https://doi.org/10.1074/jbc.M112.348250.
[71] K. Balmanno, S.J. Cook, Tumour cell survival signalling by the ERK 1/2 pathway, [94] X. Li, L. He, K.H. Che, S.F. Funderburk, L. Pan, N. Pan, M. Zhang, Z. Yue, Y. Zhao,
Cell Death Differ. 16 (2009) 368–377, https://doi.org/10.1038/cdd.2008.148. Imperfect interface of Beclin1 coiled-coil domain regulates homodimer and
[72] J.H. Kim, S.K. Hong, P.K. Wu, A.L. Richards, W.T. Jackson, J.I. Park, Raf/MEK/ heterodimer formation with Atg14l and Uvrag, Nat. Commun. 3 (2012) 662,
ERK can regulate cellular levels of LC3B and SQSTM1/p62 at expression levels, https://doi.org/10.1038/ncomms1648.
Exp. Cell Res. 327 (2014) 340–352, https://doi.org/10.1016/j. [95] A. Oberstein, P.D. Jeffrey, Y. Shi, Crystal structure of the Bcl-xl-Beclin 1 peptide
yexcr.2014.08.001. complex: Beclin 1 is a novel BH3-only protein, J. Biol. Chem. 282 (2007)
[73] U. Sivaprasad, A. Basu, Inhibition of ERK attenuates autophagy and potentiates 13123–13132, https://doi.org/10.1074/jbc.M700492200.
tumour necrosis factor-α-induced cell death in MCF-7 cells, J. Cell. Mol. Med 12 [96] Y. Zhong, Q.J. Wang, X. Li, Y. Yan, J.M. Backer, B.T. Chait, N. Heintz, Z. Yue,
(2008) 1265–1271, https://doi.org/10.1111/j.1582-4934.2008.00282.x. Distinct regulation of autophagic activity by Atg14l and rubicon associated with
[74] H. Yin, X. Yang, W. Gu, Y. Liu, X. Li, X. Huang, X. Zhu, Y. Tao, X. Gou, W. He, Beclin 1-phosphatidylinositol-3-kinase complex, Nat. Cell Biol. 11 (2009)
HMGB1-mediated autophagy attenuates gemcitabine-induced apoptosis in 468–476, https://doi.org/10.1038/ncb1854.
bladder cancer cells involving JNK and ERK activation, Oncotarget 8 (2017) [97] E. Itakura, N. Mizushima, Atg14 and uvrag: mutually exclusive subunits of
71642–71656, https://doi.org/10.18632/oncotarget.17796. mammalian beclin 1-PI3K complexes, Autophagy 5 (2009) 534–536, https://doi.
[75] G.Y. Liu, X.X. Jiang, X. Zhu, W.Y. He, Y.L. Kuang, K. Ren, Y. Lin, X. Gou, ROS org/10.4161/auto.5.4.8062.
activates JNK-mediated autophagy to counteract apoptosis in mouse [98] S. Pattingre, A. Tassa, X. Qu, R. Garuti, X.H. Liang, N. Mizushima, M. Packer, M.
mesenchymal stem cells in vitro, Acta Pharmacol. Sin. 36 (2015) 1473–1479, D. Schneider, B. Levine, Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent
https://doi.org/10.1038/aps.2015.101. autophagy, Cell 122 (2005) 927–939, https://doi.org/10.1016/j.
[76] J. Li, L. Zhao, X. Zhao, P. Wang, Y. Liu, J. Ruan, Foxo1 attenuates NaF-induced cell.2005.07.002.
apoptosis of LS8 cells through the JNK and mitochondrial pathways, Biol. Trace [99] Y. Wei, S. Pattingre, S. Sinha, M. Bassik, B. Levine, JNK1-mediated
Elem. Res. 181 (2018) 104–111, https://doi.org/10.1007/s12011-017-1015-1. phosphorylation of Bcl-2 regulates starvation-induced autophagy, Mol. Cell. 30
[77] C. Hu, M.J. Zou, L. Zhao, N. Lu, Y.J. Sun, S.H. Gou, T. Xi, Q.L. Guo, E Platinum, a (2008) 678–688, https://doi.org/10.1016/j.molcel.2008.06.001.
newly synthesized platinum compound, induces autophagy via inhibiting [100] E. Zalckvar, H. Berissi, L. Mizrachy, Y. Idelchuk, I. Koren, M. Eisenstein,
phosphorylation of mTOR in gastric carcinoma BGC-823 cells, Toxicol. Lett. 210 H. Sabanay, R. Pinkas-Kramarski, A. Kimchi, Dap-kinase-mediated
(2012) 78–86, https://doi.org/10.1016/j.toxlet.2012.01.019. phosphorylation on the BH3 domain of beclin 1 promotes dissociation of Beclin 1
[78] A. Cuadrado, A.R. Nebreda, Mechanisms and functions of p38 MAPK signalling, from Bcl-xl and induction of autophagy, EMBO Rep. 10 (2009) 285–292, https://
Biochem. J. 429 (2010) 403–417, https://doi.org/10.1042/BJ20100323. doi.org/10.1038/embor.2008.246.
[79] Q. Cui, S. Tashiro, S. Onodera, M. Minami, T. Ikejima, Oridonin induced [101] A.U. Gurkar, K. Chu, L. Raj, R. Bouley, S.H. Lee, Y.B. Kim, S.E. Dunn,
autophagy in human cervical carcinoma HeLa cells through Ras, JNK, and p38 A. Mandinova, S.W. Lee, Identification of rock1 kinase as a critical regulator of
regulation, J. Pharmacol. Sci. 105 (2007) 317–325, https://doi.org/10.1254/ beclin1-mediated autophagy during metabolic stress, Nat. Commun. 4 (2013)
jphs.fp0070336. 2189, https://doi.org/10.1038/ncomms3189.
[80] C. Zhou, J. Zhou, F. Sheng, H. Zhu, X. Deng, B. Xia, J. Lin, The heme oxygenase-1 [102] Y. Maejima, S. Kyoi, P. Zhai, T. Liu, H. Li, A. Ivessa, S. Sciarretta, D.P. Del Re, D.
inhibitor ZnPPIX induces non-canonical, Beclin 1-independent, autophagy K. Zablocki, C.P. Hsu, D.S. Lim, M. Isobe, J. Sadoshima, Mst1 inhibits autophagy
through p38 MAPK pathway, Acta Biochim. Biophys. Sin. (Shanghai). 44 (2012) by promoting the interaction between Beclin1 and Bcl-2, Nat. Med. 19 (2013)
815–822, https://doi.org/10.1093/abbs/gms064. 1478–1488, https://doi.org/10.1038/nm.3322.
[81] Y.C. Ye, L. Yu, H.J. Wang, S. Tashiro, S. Onodera, T. Ikejima, Tnfalpha-induced [103] M.C. Maiuri, G. Le Toumelin, A. Criollo, J.C. Rain, F. Gautier, P. Juin,
necroptosis and autophagy via supression of the p38-NF-κB survival pathway in E. Tasdemir, G. Pierron, K. Troulinaki, N. Tavernarakis, J.A. Hickman, O. Geneste,
l929 cells, J. Pharmacol. Sci. 117 (2011) 160–169, https://doi.org/10.1254/ G. Kroemer, Functional and physical interaction between bcl-x(l) and a BH3-like
jphs.11105fp. domain in Beclin-1, EMBO J. 26 (2007) 2527–2539, https://doi.org/10.1038/sj.
[82] A. Thyagarajan, A. Jedinak, H. Nguyen, C. Terry, L.A. Baldridge, J. Jiang, emboj.7601689.
D. Sliva, Triterpenes from ganoderma lucidum induce autophagy in colon cancer [104] A. Rasul, B. Yu, L. Zhong, M. Khan, H. Yang, T. Ma, Cytotoxic effect of evodiamine
through the inhibition of p38 mitogen-activated kinase (p38 MAPK), Nutr. Cancer in SGC-7901 human gastric adenocarcinoma cells via simultaneous induction of
62 (2010) 630–640, https://doi.org/10.1080/01635580903532390. apoptosis and autophagy, Oncol. Rep. 27 (2012) 1481–1487, https://doi.org/
[83] D. Ge, H.R. Tao, L. Fang, X.Q. Kong, L.N. Han, N. Li, Y.X. Xu, L.Y. Li, M. Yu, 10.3892/or.2012.1694.
H. Zhang, 11-methoxytabersonine induces necroptosis with autophagy through [105] Z.Y. Hu, J.P. Zhang, Effect of Beclin1 on vincristine-induced dopaminergic
AMPK/mTOR and jnk pathways in human lung cancer cells, Chem. Pharm. Bull. neurons injury in zebrafish, Acta Pharm. Sin. B 49 (2014) 843–848.
68 (2020) 244–250, https://doi.org/10.1248/cpb.c19-00851. [106] F. Sun, X. Jiang, X. Wang, Y. Bao, G. Feng, H. Liu, X. Kou, Q. Zhu, L. Jiang,
[84] S. Shimizu, A. Konishi, Y. Nishida, T. Mizuta, H. Nishina, A. Yamamoto, Y. Yang, Vincristine ablation of Sirt2 induces cell apoptosis and mitophagy via
Y. Tsujimoto, Involvement of JNK in the regulation of autophagic cell death, hsp70 acetylation in mda-mb-231 cells, Biochem. Pharmacol. 162 (2019)
Oncogene 29 (2010) 2070–2082, https://doi.org/10.1038/onc.2009.487. 142–153, https://doi.org/10.1016/j.bcp.2018.10.021.
[85] A.J. Liu, S.H. Wang, K.C. Chen, H.P. Kuei, Y.L. Shih, S.Y. Hou, W.T. Chiu, S. [107] L. Masuelli, F. Pantanella, G. La Regina, M. Benvenuto, M. Fantini, R. Mattera,
H. Hsiao, C.M. Shih, Evodiamine, a plant alkaloid, induces calcium/JNK- E. Di Stefano, M. Mattei, R. Silvestri, S. Schippa, V. Manzari, A. Modesti, R. Bei,
mediated autophagy and calcium/mitochondria-mediated apoptosis in human Violacein, an indole-derived purple-colored natural pigment produced by
glioblastoma cells, Chem. Biol. Interact. 205 (2013) 20–28, https://doi.org/ janthinobacterium lividum, inhibits the growth of head and neck carcinoma cell
10.1016/j.cbi.2013.06.004. lines both in vitro and in vivo, Tumor Biol. 37 (2016) 3705–3717, https://doi.
[86] X. Yang, J. Gao, J. Guo, Z. Zhao, S.L. Zhang, Y. He, Anti-lung cancer activity and org/10.1007/s13277-015-4207-3.
inhibitory mechanisms of a novel calothrixin a derivative, Life Sci. 219 (2019) [108] Y. Shang, Q. Wang, J. Li, Q. Zhao, X. Huang, H. Dong, H. Liu, Y. Zhang, J. Zhang,
20–30, https://doi.org/10.1016/j.lfs.2018.12.052. R. Gui, X. Nie, The acetone indigo red dehydrating agent IF203 induces HepG2
[87] T. Utaipan, A. Athipornchai, A. Suksamrarn, S. Chunsrivirot, W. Chunglok, cell death through cell cycle arrest, autophagy and apoptosis, Onco Targets Ther.
Isomahanine induces endoplasmic reticulum stress and simultaneously triggers 13 (2020) 473–486, https://doi.org/10.2147/OTT.S232594.
p38 MAPK-mediated apoptosis and autophagy in multidrug-resistant human oral [109] H. Steinbrenner, H. Sies, Protection against reactive oxygen species by
squamous cell carcinoma cells, Oncol. Rep. 37 (2017) 1243–1252, https://doi. selenoproteins, Biochim. Biophys. Acta 2009 (1790) 1478–1485, https://doi.org/
org/10.3892/or.2017.5352. 10.1016/j.bbagen.2009.02.014.
[88] L. Hu, T. Zhang, D. Liu, G. Guan, J. Huang, P. Proksch, X. Chen, W. Lin, [110] A. Rahal, A. Kumar, V. Singh, B. Yadav, R. Tiwari, S. Chakraborty, K. Dhama,
Notoamide-type alkaloid induced apoptosis and autophagyviaa p38/JNK Oxidative stress, prooxidants, and antioxidants: The interplay, BioMed. Res. Int.
signaling pathway in hepatocellular carcinoma cells, RSC Adv. 9 (2019) 2014 (2014), 761264, https://doi.org/10.1155/2014/761264.
19855–19868, https://doi.org/10.1039/c9ra03640g. [111] R. Scherz-Shouval, Z. Elazar, Regulation of autophagy by ros: physiology and
[89] A. Abe, H. Yamada, S. Moriya, K. Miyazawa, The β-carboline alkaloid harmol pathology, Trends Biochem. Sci. 36 (2011) 30–38, https://doi.org/10.1016/j.
induces cell death via autophagy but not apoptosis in human non-small cell lung tibs.2010.07.007.
cancer A549 cells, Biol. Pharm. Bull. 34 (2011) 1264–1272, https://doi.org/ [112] G. Filomeni, D. De Zio, F. Cecconi, Oxidative stress and autophagy: the clash
10.1248/bpb.34.1264. between damage and metabolic needs, Cell Death Differ. 22 (2015) 377–388,
[90] N. Ishimwe, P. Wei, M. Wang, H. Zhang, L. Wang, M. Jing, L. Wen, Y. Zhang, https://doi.org/10.1038/cdd.2014.150.
Autophagy impairment through lysosome dysfunction by brucine induces [113] J. Yang, L.J. Wu, S. Tashino, S. Onodera, T. Ikejima, Reactive oxygen species and
immunogenic cell death (ICD), J. Tradit. Chin. Med. 48 (2020) 1915–1940, nitric oxide regulate mitochondria-dependent apoptosis and autophagy in
https://doi.org/10.1142/s0192415x20500962. evodiamine-treated human cervix carcinoma hela cells, Free Radic. Res. 42
[91] J. Chen, Q. Guo, J. Zhang, Z. Yin, W. Song, B. He, Y. Zhang, W. Zhang, L. Chen, (2008) 492–504, https://doi.org/10.1080/10715760802112791.
Chaetoglobosin G inhibits proliferation, autophagy and cell cycle of lung cancer [114] T. Itoh, R. Tsuzuki, T. Tanaka, M. Ninomiya, Y. Yamaguchi, H. Takenaka,
cells through EGFR/MEK/ERK signaling pathway, Pharmazie 75 (2020) 642–645, M. Ando, Y. Tsukamasa, M. Koketsu, Reduced scytonemin isolated from Nostoc
https://doi.org/10.1691/ph.2020.0750. commune induces autophagic cell death in human t-lymphoid cell line jurkat

11
M.-L. Luo et al. Biomedicine & Pharmacotherapy 149 (2022) 112827

cells, Food Chem. Toxicol. 60 (2013) 76–82, https://doi.org/10.1016/j. attenuation of lysosomal acidification, J. Nat. Prod. 83 (2020) 1432–1439,
fct.2013.07.016. https://doi.org/10.1021/acs.jnatprod.9b00856.
[115] W. Bi, Y. Bi, P. Li, S. Hou, X. Yan, C. Hensley, C.E. Bammert, Y. Zhang, K. [125] T. Donia, M.N. Gerges, T.M. Mohamed, Anticancer effects of combination of
M. Gibson, J. Ju, L. Bi, Indole alkaloid derivative B, a novel bifunctional agent Indole-3-carbinol and hydroxychloroquine on ehrlich ascites carcinoma via
that mitigates 5-fluorouracil-induced cardiotoxicity, ACS Omega 3 (2018) targeting autophagy and apoptosis, Nutr. Cancer (2021) 1–17, https://doi.org/
15850–15864, https://doi.org/10.1021/acsomega.8b02139. 10.1080/01635581.2021.1960388.
[116] N. Meng, X. Mu, X. Lv, L. Wang, N. Li, Y. Gong, Autophagy represses fascaplysin- [126] K. Fang, S.P. Chen, C.W. Lin, W.C. Cheng, H.T. Huang, Ellipticine-induced
induced apoptosis and angiogenesis inhibition via ros and p8 in vascular apoptosis depends on Akt translocation and signaling in lung epithelial cancer
endothelia cells, Biomed. Pharmacother. 114 (2019), 108866, https://doi.org/ cells, Lung Cancer 63 (2009) 227–234, https://doi.org/10.1016/j.
10.1016/j.biopha.2019.108866. lungcan.2008.05.026.
[117] J. Shin, P62 and the sequestosome, a novel mechanism for protein metabolism, [127] J.P. Wang, Y.C. Yu, S.P. Chen, H.C. Liang, C.W. Lin, K. Fang, The collective
Arch. Pharm. Res. 21 (1998) 629–633, https://doi.org/10.1007/bf02976748. nuclear migration of p53 and phosphorylated s473 of Akt during ellipticine-
[118] S. McManus, S. Roux, The adaptor protein p62/SQSTM 1 in osteoclast signaling mediated apoptosis in human lung epithelial cancer cells, Mol. Cell. Biochem. 407
pathways, J. Mol. Signal. 7 (2012) 1, https://doi.org/10.1186/1750-2187-7-1. (2015) 123–133, https://doi.org/10.1007/s11010-015-2460-9.
[119] M.L. Seibenhener, J.R. Babu, T. Geetha, H.C. Wong, N.R. Krishna, M.W. Wooten, [128] X. Sun, L. Li, H.G. Ma, P. Sun, Q.L. Wang, T.T. Zhang, Y.M. Shen, W.M. Zhu, X. Li,
Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in Bisindolylmaleimide alkaloid bma-155cl induces autophagy and apoptosis in
ubiquitin proteasome degradation, Mol. Biol. Cell. 24 (2004) 8055–8068, https:// human hepatocarcinoma Hepg-2 cells through the Nf-kappab p65 pathway, Acta
doi.org/10.1128/MCB.24.18.8055-8068.2004. Pharmacol. Sin. 38 (2017) 524–538, https://doi.org/10.1038/aps.2016.171.
[120] L.J. Kraft, J. Dowler, P. Manral, A.K. Kenworthy, Size, organization, and dynamics [129] W.S. Kong, F.X. Shen, R.F. Xie, G. Zhou, Y.M. Feng, X. Zhou, Bufothionine induces
of soluble SQSTM1 and lc3-SQSTM1 complexes in living cells, Autophagy 12 autophagy in H22 hepatoma-bearing mice by inhibiting Jak2/stat3 pathway, a
(2016) 1660–1674, https://doi.org/10.1080/15548627.2016.1199299. possible anti-cancer mechanism of cinobufacini, J. Ethnopharmacol. 270 (2021),
[121] K.I. Lee, M.J. Kim, H. Koh, J.I. Lee, S. Namkoong, W.K. Oh, J. Park, The anti- 113848, https://doi.org/10.1016/j.jep.2021.113848.
hypertensive drug reserpine induces neuronal cell death through inhibition of [130] S. Song, S. Kim, E.R. El-Sawy, C. Cerella, B. Orlikova-Boyer, G. Kirsch, C. Christov,
autophagic flux, Biochem. Biophys. Res. Commun. 462 (2015) 402–408, https:// M. Dicato, M. Diederich, Anti-leukemic properties of aplysinopsin derivative EE-
doi.org/10.1016/j.bbrc.2015.04.145. 84 alone and combined to BH3 mimetic A-1210477, Mar. Drugs 19 (2021),
[122] X. Xiao, M. Xu, C. Yang, Y. Yao, L.N. Liang, P. Ed Chung, Q. Long, E. Zacksenhaus, https://doi.org/10.3390/md19060285.
Z. He, S. Liu, Y. Ben-David, Novel racemosin b derivatives as new therapeutic [131] L. Galluzzi, M. De Santi, R. Crinelli, C. De Marco, N. Zaffaroni, A. Duranti,
agents for aggressive breast cancer, in: Bioorg. Med. Chem., 26, 2018, G. Brandi, M. Magnani, Induction of endoplasmic reticulum stress response by the
pp. 6096–6104, https://doi.org/10.1016/j.bmc.2018.11.014. indole-3-carbinol cyclic tetrameric derivative ctet in human breast cancer cell
[123] T. Ohashi, Y. Nakade, M. Ibusuki, R. Kitano, T. Yamauchi, S. Kimoto, T. Inoue, lines, PLoS One 7 (2012) 43249, https://doi.org/10.1371/journal.pone.0043249.
Y. Kobayashi, Y. Sumida, K. Ito, H. Nakao, K. Umezawa, M. Yoneda, Conophylline [132] Y. Sasazawa, N. Sato, K. Umezawa, S. Simizu, Conophylline protects cells in
inhibits high fat diet-induced non-alcoholic fatty liver disease in mice, PLoS One cellular models of neurodegenerative diseases by inducing mammalian target of
14 (2019), e0210068, https://doi.org/10.1371/journal.pone.0210068. rapamycin (mTOR)-independent autophagy, J. Biol. Chem. 290 (2015)
[124] Y. Zhang, X. Ding, Y.X. Yuan, L.L. Guo, X.J. Hao, Cytotoxic monoterpenoid indole 6168–6178, https://doi.org/10.1074/jbc.M114.606293.
alkaloids from tabernaemontana corymbosa as potent autophagy inhibitors by the

12

You might also like