Sunteți pe pagina 1din 10

Critical Reviews in Oncology/Hematology 112 (2017) 2130

Contents lists available at ScienceDirect

Critical Reviews in Oncology/Hematology


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

The roles of reactive oxygen species (ROS) and autophagy in the


survival and death of leukemia cells
Yong-Feng Chen a,b, , Hao Liu c , Xin-Jing Luo a,b , Zhiqiang Zhao a,b , Zhen-You Zou a,b,d ,
Jing Li e , Xiao-Jing Lin f , Yong Liang b,
a
Department of Basic Medical Sciences, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China
b
Institute of Tumor, School of Medicine of Taizhou University, Taizhou 318000, Zhejiang, China
c
School of Pharmacy, Shanghai Jiaotong University, Shanghai 200240, China
d
Biochemistry Department of Purdue University, West Lafayette, IN 47906, USA
e
Department of Histology and Embryology, North SiChuan Medical College, Nanchong 637000, Sichuan, China
f
Department of Hematology, the Afliated Hospital of Guiyang Medical College, Guiyang 550004, China

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
2. The role of ROS in hematopoiesis and the pathogenesis of leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
3. The role of autophagy in hematopoiesis and the pathogenesis of leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
4. ROS autophagy interaction implicated in the evolution of MDS to AML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5. Exploiting ROS for overcoming drug resistance in leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
6. Targeting autophagy to overcome drug resistance in leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
7. ROS-induced autophagy as a therapeutic strategy for treatment of leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
8. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Conict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28

a r t i c l e i n f o a b s t r a c t

Article history: As a clonal disease of hematopoietic stem cells (HSCs), the etiology and pathogenesis of leukemia is not
Received 17 November 2015 fully understood. Recent studies suggest that cellular homeostasis plays an essential role in maintaining
Received in revised form the function of HSCs because dysregulation of cellular homeostasis is one of the major factors underlying
27 November 2016
the malignant transformation of HSCs. Reactive oxygen species (ROS) and autophagy, key factors regu-
Accepted 6 February 2017
lating cellular homeostasis, are commonly observed in the human body. Autophagy can be induced by
ROS through a variety of signaling pathways, and conversely inhibits ROS-induced damage to cells and
Keywords:
tissues. ROS and autophagy coordinate to maintain cellular homeostasis. Previous studies have demon-
Reactive oxygen species (ROS)
Autophagy
strated that both of ROS and autophagy play important roles in the development of leukemia and are
Leukemia closely involved in drug resistance in leukemia. Interference with cellular homeostasis by promoting pro-
Hematopoiesis grammed leukemia cell death via ROS and autophagy has been veried to be an efcient technique in the
Homeostasis treatment of leukemia. However, the critical roles of ROS and autophagy in the development of leukemia
Programmed cell death are largely unknown. In this review, we summarize the roles of ROS and autophagy in the pathogenesis
Drug resistance of leukemia, which may allow the identication of novel targets and drugs for the treatment of leukemia
based on the regulation of HSCs homeostasis through ROS and autophagy.
2017 Elsevier B.V. All rights reserved.

1. Introduction

Corresponding author at: School of Medicine of Taizhou University, Taizhou, Reactive oxygen species (ROS) are a class of oxygen metabo-
318000, Zhejiang, China. lites and oxygen-containing materials that are derived from oxygen
Corresponding author. metabolites, and they include all peroxide and oxygen-free radi-
E-mail addresses: cyfeng@tzc.edu.cn (Y.-F. Chen), liangytzc@163.com (Y. Liang).

http://dx.doi.org/10.1016/j.critrevonc.2017.02.004
1040-8428/ 2017 Elsevier B.V. All rights reserved.
22 Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130

cals. ROS are chemically more active than oxygen, tightly associated lization of HSCs (Ludin et al., 2014; Juntilla et al., 2010); however,
with cellular homeostasis, and can signicantly affect cellular higher levels of ROS have toxic effects on HSCs (Shao et al., 2011).
homeostasis and metabolism (Nogueira and Hay, 2013a). In normal HSCs reside mainly in the bone marrow in the quiescent state. Com-
concentrations, ROS serve as important second messengers that pared with other types of cells, HSCs are more susceptible to ROS
are involved in a variety of signal transduction events that regu- (Jang and Sharkis, 2007; Richardson et al., 2015). Increased levels
late the growth, proliferation, and differentiation of cells (Landry of ROS can result in DNA damage in HSCs, leading to dysfunctions
and Cotter, 2014; Kobashigawa et al., 2015). Redox dysregulation such as abnormal proliferation and differentiation (Yahata et al.,
causes an excessive generation and accumulation of ROS, leading 2011). In the bone marrow microenvironment, many cells, such as
to oxidative damage to cells and tissues, which is involved in the -smooth muscle actin (SMA)+ macrophages and mesenchymal
development of cancers and many other human diseases (Montero stromal cells (MSCs), are involved in maintaining the balance of ROS
and Jassem, 2011; Nogueira and Hay, 2013b). in stem cells (Fig. 1) (Ludin et al., 2014). In addition, the deletion of
Autophagy, which involves lysosomal-dependent degradation a number of genes such as FoxO3 or Atm can increase ROS levels
of macromolecules and organelles in cells, plays an essential role in and inhibit HSCs self-renewal and hematopoiesis, suggesting that
maintaining cellular homeostasis and is involved in the pathogen- these genes are involved in ROS production and the regulation of
esis of numerous diseases (Chen et al., 2014a). A number of signal hematopoiesis (Zhou et al., 2013).
transduction pathways are related to the function and coordination Under normal physiological conditions, the generation and
between ROS and autophagy in cells in response to cellular stress. degradation of ROS in the body maintain in a state of equilibrium.
ROS and autophagy interact to maintain cellular homeostasis. ROS An imbalance between oxidant and antioxidant systems results
can induce autophagy, but autophagy serves as a buffer system to in the over-production and accumulation of ROS. Previous studies
control the level of ROS in cells and reduce their toxic effects (Li have shown that ROS over-production due to redox dysregulation
et al., 2015). Redox imbalances and/or autophagy dysregulations plays an important role in the proliferation, survival, differentia-
affect the proliferation and differentiation of hematopoietic stem tion, immune escape, and epigenetic changes observed in leukemia
cells (HSCs), leading to the development of hematological malig- cells (Lewandowski et al., 2010). Devi et al. observed increased
nancies in some degree (Naka et al., 2008; Evangelisti et al., 2015). levels of superoxide anion and hydrogen peroxide generated by
Recent progress in leukemia has revealed that ROS and autophagy polymorphonuclear leukocytes in the peripheral blood of untreated
are strongly associated with the development of this disease. In leukemia patients (Devi et al., 2000). Interestingly, the changes in
addition, they are also potentially involved in the mechanism of superoxide anion and hydrogen peroxide levels in the peripheral
drug resistance of leukemia cells. It has been reported that the blood were not associated with the leukemia types, suggesting that
disruption of cellular homeostasis through ROS and autophagy increased ROS is a common feature of all leukemia types (Devi
is an effective approach to circumvent the development of drug et al., 2000). It has been reported that high levels of ROS can lead
resistance in leukemia cells (Evangelisti et al., 2015; Udensi and to genomic instability and are associated with the development
Tchounwou, 2014; She et al., 2007; Ekiz et al., 2012; Torgersen and progression of leukemia (Popp and Bohlander, 2010). Rassool
and Simonsen, 2013). However, the roles of ROS and autophagy et al. reported that DNA damage and error-prone repair of damaged
in the development and treatment of leukemia remain unclear. DNA were consistent with the increased levels of ROS detected in a
In particular, the signal transduction pathways involving ROS and myeloid leukemic disease progression model (Rassool et al., 2007).
autophagy in the maintenance of cellular homeostasis are largely Zhou et al. found that the serum level of 8-oxo-2 -deoxyguanosine
unknown. Elucidation of the molecular mechanisms underlying (8-oxo-dG), a biomarker of oxidative damage to DNA, was signif-
ROS and autophagy in the pathogenesis of leukemia may provide icantly higher in acute myeloid leukemia (AML) patients than in
novel targets and drugs for the treatment of this disease. normal controls (Zhou et al., 2010). In addition, Collado et al. found
that the levels of 8-oxo-dG were signicantly higher in the lympho-
cytes of patients with CLL compared with normal controls (Collado
2. The role of ROS in hematopoiesis and the pathogenesis et al., 2012). Taken together, these results suggest that oxidative
of leukemia DNA damage caused by ROS is closely associated with the develop-
ment of leukemia.
ROS can be classied as exogenous or endogenous ROS accord- To date, the precise mechanisms underlying ROS overproduc-
ing to their origins. Exogenous ROS are generated in the body by tion remain largely unknown. Recent studies suggest that increased
external sources such as radiation and drugs, and endogenous ROS production of ROS in AML, chronic myelogenous leukemia (CML),
are mainly the products of aerobic metabolism in cells (Lee et al., and promyelocytic leukemia (PL) is mainly attributed to the consti-
2013). ROS can be generated by a number of approaches, including tutive activation of NOXs (Hole et al., 2013; Singh et al., 2012; Dong
mitochondria, the NADPH oxidase system (NOXs), and the xanthine et al., 2004). Hole et al. also found that elevated ROS in AML cells
oxidase system, among which NOXs are the most important source was associated with reduced levels of glutathione (GSH) and deple-
of ROS (Libik-Konieczny et al., 2015). NOXs are located in the cyto- tion of antioxidant defense proteins (Hole et al., 2013). In addition,
plasmic membrane and can be quickly activated by bacterial lipid reduced antioxidant levels were also identied in other types of
polysaccharides, TNF-, or IL-1 to produce ROS, a second messen- leukemia (Battisti et al., 2008; Oltra et al., 2001; Silber et al., 1992),
ger molecule that is involved in a variety of signal transduction suggesting that the imbalance between oxidant and antioxidant
pathways (Dickinson and Chang, 2011). systems is a major cause of the overproduction of ROS. However,
Normal levels of ROS are essential for maintaining the phys- improved antioxidant responses have also been observed in differ-
iological functions of cells by regulating their proliferation and ent types of leukemia and may represent adaptive responses that
differentiation, and for controlling immune responses and cellu- protect cells against oxidative stress damage (Devi et al., 2000).
lar inammation (Owusu-Ansah and Banerjee, 2009; Kwak et al., These inconsistent results may be explained by the heterogeneous
2015). Excess ROS can cause oxidative stress, leading to oxidative nature of leukemia, for which the oxidant and antioxidant levels
damage to organelles, proteins, lipids, and DNA, disruption of cellu- detected in different type of leukemia or the same type at differ-
lar structures and functions, or cell death (Yorimitsu and Klionsky, ent disease stages are distinct. However, this hypothesis requires
2007). ROS regulate the function of HSCs in a concentration- further investigation.
dependent manner. At normal levels, ROS serve as a signaling As the major source of ROS, NOXs improve both the survival
molecule to control the proliferation, differentiation, and mobi- and proliferation of leukemia cells by increasing the production of
Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130 23

Fig. 1. Environmental factors maintain the balance of ROS in stem cells. Mesenchymal stromal cells (MSCs) and -smooth muscle actin (SMA)+ macrophages cooperate to
maintain low levels of ROS in HSCs. The SMA+ macrophages contain cyclooxygenase-2 (COX-2), which produces prostaglandin E2 (PGE2) to reduce intracellular ROS levels
in stem cells. PGE2 also promotes CXCL12 production from MSCs. In addition, stem cells are connected to the MSCs via connexin gap junctions that transfer ROS to reduce
ROS levels in stem cells. This gure was adapted from (Ludin et al., 2014).

ROS. However, the underlying mechanism is not fully understood. etic stem/progenitor cells, ROS accumulation in mitochondria,
Singh et al. found that heme oxygenase 1 (HO-1), a cytoprotective DNA damage, and myeloproliferation (Mortensen et al., 2011). In
protein that plays a critical role against oxidative stress, was up- vitro experiments had shown that Atg7 knockout caused a seri-
regulated in CML cells by the NOXs components Rac1 and p47phox ous decline in the colony-forming ability of HSCs (Mortensen
(Singh et al., 2012). Our previous study also showed that HO-1 was et al., 2011). Similarly, the autophagy inhibitor 3-methyladenine
aberrantly overexpressed in a majority of AML patients, especially (3-MA) or Atg5 siRNA suppressed colony formation in HSCs (Salemi
patients with acute monocytic leukemia (M5) and leukocytosis (Lin et al., 2012). Taken together, these results suggest that autophagy
et al., 2015). Our results further demonstrated that HO-1 inhibited plays an important role in maintaining homeostasis, self-renewal,
AML cell apoptosis by activating the JNK/c-JUN signaling path- and differentiation in HSCs; however, the underlying mecha-
way (Lin et al., 2015). Aurelius et al. recently found that the ROS nisms require further investigation. The autophagy-related genes
generated from NOXs in leukemia cells promoted NK and T cell involved in hematopoiesis are shown in Table 1.
apoptosis, allowing leukemia cells to escape from antileukemic It is recently identied that autophagy is associated with
lymphocytes (Aurelius et al., 2012). These results suggest that the hematopoietic dysplasia and involved in the occurrence and devel-
NOXs-ROS pathway is a potentially powerful target for the treat- opment of certain types of leukemia. Robust evidence exists that
ment of leukemia. autophagy involved in the development of BCR-ABL-mediated
leukemia. The BCR/ABL rearrangement t(9;22)(q34;q11) (Ph chro-
mosome) represents the hallmark of CML and can also be detected
3. The role of autophagy in hematopoiesis and the
in ALL (Chiaretti et al., 2014). The BCR-ABL protein has been shown
pathogenesis of leukemia
to induce ROS production and DNA damage in CML cells (Sattler
et al., 2000; Benhar et al., 2001). In addition, the BCR-ABL protein,
Under normal physiological conditions, autophagy is main-
which possesses extremely high levels of tyrosine kinase activity,
tained at a low level; however, autophagy can be activated by
exogenous stimuli such as oxidative stress, hypoxia, nutritional
deciencies, and infection. Intracellular soluble proteins, long-lived Table 1
Potential roles of autophagy in hematopoiesis.
proteins, and damaged organelles are transported to the lysosome
for degradation. The degraded products, including free fatty acids, Hematopoiesis process Autophagy-related
amino acids, and nucleotides, will be reused by cells. Excessive acti- genes involved in
hematopoiesis
vation of autophagy may induces autophagic cell death (Clarke and
Puyal, 2012). HSCs self-renewal ATG7, IRGM, MTOR
Although the mechanism underlying autophagy has been exten- HSCs quiescence ATG7, IRGM,
RB1CC1/FIP200
sively studied in recent years, the role and regulation of autophagy HSCs progenitor ATG7, RB1CC1/FIP200
in hematopoiesis remains unknown. Recently, Mortensen et al. differentiation/remodeling
reported that conditional knockout of the autophagy-related genes, Lineage-restricted progenitor ATG3, ATG5, ATG7,
Atg7 or FIP200, in the hematopoietic system of mice led to a maintenance ULK1, BNIP3L/NIX,
PIK3C3/VPS34
functional deciency of HSCs, signicant decrease in hematopoi-
24 Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130

can phosphorylate itself and many other substrates, activating the tion of intracellular organelles and proteins to provide energy and
RAS/MAPK, PI3K/AKT, and JAK/STAT signaling pathways. Activation nutrients for tumor cell growth. In addition, autophagy can remove
of these pathways has extensive effects on the growth, differenti- or repair ROS-damaged DNA and proteins, reducing cytotoxicity
ation, and survival of hematopoietic cells, including an increase in and protecting cells (Sun et al., 2010; Lisanti et al., 2010; He and
proliferation and inhibition of apoptosis, thus promoting the devel- Klionsky, 2009). However, autophagy may fail to remove excess
opment of CML (Samanta et al., 2011; Stella et al., 2013). Altman ROS when ROS levels exceed the autophagy capacity, resulting in
et al. reported that autophagy was the primary determinant of the excess autophagy and autophagic cell death (Dadakhujaev et al.,
survival of cells expressing BCR-ABL, whereas the basal levels of 2009).
autophagy in BCR-ABL-expressing cells were low (Altman et al., Mitochondria are the main organelles responsible for ROS pro-
2011). The authors also found that knockout of the autophagy- duction and degradation, but they are also highly sensitive to
related gene Atg3 or autophagy inhibitors signicantly induced ROS damage. Mitochondrial damage or dysfunction reduces the
apoptosis (Altman et al., 2011). Transplantation of BCR-ABL cells production of ATP and increases the production of ROS, increas-
into mice caused leukemia 20 days after transplantation (Altman ing oxidative damage to cells (Gobe and Crane, 2010). To date,
et al., 2011). These results suggest that autophagy plays an essential numerous studies have suggested that mitochondrial dysfunc-
role in the development of BCR-ABL-mediated leukemia and that tion is associated with the development of Parkinsons disease,
the survival of BCR-ABL cells might depends mainly on autophagy. Alzheimers disease, diabetes, and many other diseases (de Moura
Different from its role in CML, autophagy may play a distinct role et al., 2010; Jung and Lee, 2010; Pallard et al., 2010). Abnormal
in the development of AML. Waston et al. declared that if there was mitochondria, such as increased numbers of mitochondria with an
autophagy defect in the process of bone marrow hematopoiesis, a irregular morphology displaying a destroyed mitochondrial matrix
large number of bone marrow progenitor cells would accumulate, and membrane and disorganized cristae, have also been observed
which would lead to the dysfunctions of hematopoietic stem cell in leukemia cells (Schumacher et al., 1975; Szekely et al., 1976;
and myeloid differentiation, and severe myeloid invasion, a symp- Herrera-Goepfert et al., 1986; Iwama and Eguchi, 1986). There-
tom extremely similar to that in acute myeloid leukemia. Irregular fore, it has been proposed that the oxygen-peroxide features of
myeloid proliferation occurred in the bone marrow of autophagy- mitochondria are involved in leukemogenesis (Lyu et al., 2008).
related gene (Atg) 7 knockout mice, which was similar to the MDS was recognized as hematopoietic dysfunction due to
process of myelody splastic syndrome (MDS) to AML in human. abnormal proliferation and differentiation of HSCs. Approximately
It hints that autophagic defects may be associated with the devel- one third of MDS patients will nally develop into AML. Interest-
opment of MDS to AMI (Watson et al., 2011a). Hu et al. reported ingly, elevated ROS levels accompanied by mitochondrial damage
that the expression of BECLIN-1 and the number of autophagic vac- was observed in mononuclear bone marrow cells of MDS patients
uoles in bone marrow mononuclear cells (BMMNC) from patients (Farquhar and Bowen, 2003). As mentioned before, mitochondrial
with MDS progression and patients with MDS-transformed AML damage is a direct cause of many human diseases, and mitochon-
were gradually declining (Hu et al., 2015). Besides, plenty of rela- drial autophagy is an important regulatory mechanism of cells
tive studies had obtained the analogous results (Wan et al., 2013; via the removal of damaged mitochondria and maintaining their
Zare-Abdollahi et al., 2016). Totally, it suggests that autophagy may homeostasis. When the cells are autophagy defects, it will lead
be closely related to the progression of AML. to intracellular damage and a large number of aggregation in the
aged mitochondria, thereby promoting the development of tumor.
Boultwood et al. conducted quantitative analyses of mitochon-
4. ROS autophagy interaction implicated in the evolution drial DNA (mtDNA) in the peripheral blood of AML patients and
of MDS to AML healthy individuals. They found a signicantly greater quantity of
mtDNA in AML patients compared with healthy individuals, sug-
Recent studies show that a variety of cell homeostasis mecha- gesting that mitochondrial autophagy defect may exist in AML
nisms work together to reduce ROS damage to cells and maintain cells, leading to an increase in the number of mitochondria, and
cell survival. Autophagy is one of the major mechanisms maintain- that the mitochondrial autophagy defect may involved in the
ing cellular homeostasis. ROS and autophagy play important roles development of AML (Boultwood et al., 1996). Houwerzijl et al.
in stress response in cells through a number of complicated sig- reported that mitophagy activity in erythroid precursors was signif-
naling pathways and molecules (Underwood et al., 2010; Xu et al., icantly increased in patients with AML-high-risk myelodysplastic
2006; Cao et al., 2009; Kongsuphol et al., 2009; Chen et al., 2014b). syndrome (MDS) compared with those with AML-low-risk MDS,
A variety of stresses, such as nutritional deprivation, hypoxia, suggesting that mitophagy is an important protective mechanism
ischemia-reperfusion, and cellular stress, can increase intracellu- for the degradation of abnormal mitochondria and inhibition of ROS
lar ROS production, which subsequently induces autophagy (Essick accumulation in cells (Houwerzijl et al., 2009). Some investigators
and Sam, 2010). This sequence of events has been described have proposed that high cellular levels of ROS cause DNA dam-
in a number of diseases, such as diabetes, rheumatoid arthri- age and increase the risk of developing AML. Therefore, functional
tis, heart failure, neurodegenerative diseases, cancer, and cystic autophagy may be an important mechanism for the prevention of
brosis (Essick and Sam, 2010). In addition, a variety of exoge- MDS into AML (Watson et al., 2011b).
nous substances can induce autophagy by increasing ROS levels.
The pathways involved in ROS-induced autophagy are affected by
numerous factors, such as cell types, different stimuli, and the 5. Exploiting ROS for overcoming drug resistance in
experimental environment. Regulation of autophagy by ROS is leukemia
shown in Fig. 2.
Recent studies had demonstrated that some types of tumor cells, Great progress have been achieved in the treatment of leukemia;
such as breast cancer, liver cancer, stomach cancer, colorectal can- however, the emergence of drug resistance often leads to treatment
cer, and cervical cancer cells, possess higher levels of autophagy failure. Therefore, understanding the mechanisms underlying drug
activity (Ahn et al., 2007; Tang et al., 2009; Karantza-Wadsworth resistance in leukemia is critically important to predict treatment
and White, 2007; Sun et al., 2010). Tumor cells often reside in a outcomes. Drug resistance in leukemia involves complicated mech-
hypoxic environment with nutritional deciencies. Autophagy may anisms, such as enhanced drug efux, proto-oncogene mutations,
be an important tumor cell survival mechanism via the degrada- the amplication of drug resistance genes, increased activities of
Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130 25

Fig. 2. Regulation of autophagy by reactive oxygen species (ROS). ROS are mainly produced in mitochondria. Increased ROS production caused by nutrient starvation, drugs,
and radiation, regulates autophagy via several signaling pathways such as the AMPK-mTOR and PI3K-Akt-mTOR pathways (Jung and Lee, 2010). In addition, increased ROS
production also leads to a redox imbalance in cells through the release of GSH into the environment via MRP1, which decreases the GSH/GSSG ratio and oxidized thiols and
causes autophagy (Pallard et al., 2010).
AMPK: AMP-activated protein kinase; ATG4: autophagy-related gene 4; Bcl-2: B-cell lymphoma-2; Beclin-1: Bcl-2 interacting coiled-coil protein-1; ERk 1/2: extracellular
signal-regulated kinase1/2; FOXO: Forkhead box-containing protein O subfamily; GSH: reduced glutathione; GSSG: oxidized glutathione; JNK: cJun n-terminal kinase; LC3-II:
microtubule associate protein 1 light chain 3-II; MRP1: multidrug resistance protein 1; mTOR: mammalian target of rapamycin; PI3K: phosphatidylinositol 3-kinase; PKB/Akt:
protein kinase B; PTEN: phosphatase and tensin homologue deleted on chromosome10; TSC1/2: tuberous sclerosis complex 1/2; Ulk1/2: unc-51-like kinase 1/2.
26 Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130

drug-metabolizing enzymes, and inhibition of apoptosis (Frame, and increased autophagy in T-cell acute lymphoblastic leukemia
2007; Chauncey, 2001). In recent years, the role of ROS in tumor (T-ALL) cells, and the autophagy inhibitor chloroquine signi-
resistance has received much attention. The elevation of drug cantly improved apoptosis in T-ALL cells induced by triciribine
resistance genes expression was reported as one of the poten- (Evangelisti et al., 2011). In addition, inhibition of autophagy has
tial mechanisms of ROS affecting tumor resistance (Hwang et al., been shown to restore the effects of Dasatinib on apoptosis in CLL
2007; Tien Kuo and Savaraj, 2006). Besides, ROS may also affect cells (Amrein et al., 2011). Histone deacetylase inhibitors inhibit
tumor resistance through a variety of ways. Yi et al. found that autophagy and improve apoptosis in myeloid leukemia cells by
the intracellular ROS level determined the sensitivity of leukemia increasing ROS production, and these phenomena are reversed by
cells to arsenic trioxide (Yi et al., 2002). For example, a higher the activation of autophagy (Stankov et al., 2014). Taken together,
level of intracellular ROS in leukemia cells was associated with an these results suggest that autophagy is a self-protective mecha-
elevated sensitivity to arsenic trioxide. Zhou et al. also reported nism of leukemia cells and that targeting autophagy is an efcient
that CLL cells with a higher basal level of ROS were more sensi- approach to resolve the challenge of drug resistance in the treat-
tive to the chemotherapeutic agent 2-methoxyestradiol than CLL ment of leukemia.
cells with a lower basal level of ROS (Zhou et al., 2003). All these Recent studies have identied that numerous signaling
studies indicated that ROS level in leukemia cells might be tightly molecules and pathways are involved in the regulation of
associated with related cell resistance, low ROS level with greater autophagy. Some of these signaling molecules, such as High mobil-
resistance, and high ROS level followed by weaker resistance. Zhou ity group protein 1 (HMGB1), miRNAs, and p53, are directly or
et al. proved that the raise of ROS production improved the anti- indirectly involved in the drug resistance of leukemia cells. HMGB1
leukemic activity of 2-methoxyestradiol (Zhou et al., 2003). In is a nuclear DNA-binding protein and a member of the damage-
addition, some studies have also demonstrated that abrogation of associated molecular patterns (DAMPs). HMGB1 has been shown
the redox balance in leukemia cells by increasing the production of to be a positive regulator of autophagy that is released from cells
ROS is an efcient strategy to overcome drug resistance in leukemia under stress conditions such as injury and infections (Yu et al.,
(Ruvolo et al., 2010; Tonino et al., 2011; Kim et al., 2014; Changchien 2012a). Liu et al. reported that leukemia cells secreted HMGB1 in
et al., 2015). response to the toxic effects of chemotherapeutic agents, which
According to reports in the literature, the anti-leukemic prop- activated autophagy to protect the cells (Liu et al., 2011). Anti-
erties of a variety of chemotherapy drugs that are used to treat HMGB1 antibody improved the sensitivity of leukemia cells to
leukemia, such as doxorubicin, vincristine, and arabinoside, have chemotherapeutic agents, suggesting that HMGB1 is associated
been attributed to the generation of ROS in leukemia cells(Mizutani with the drug resistance of leukemia cells. In addition, blocking
et al., 2005; Groninger et al., 2002; Romano et al., 2000; Kanno et al., the PI3KMEKERK pathway inhibited leukemia cell autophagy
2004; Guzman et al., 2005; Yang et al., 2006; Inoue et al., 1994; Chen induced by HMGB1 (Liu et al., 2011). miRNAs are a class of reg-
et al., 2005; Yaseen et al., 2012; Ka et al., 2003; Efferth et al., 2007). ulatory RNA that plays an important role in the development,
Mizutani et al. compared the sensitivity to doxorubicin between metastasis, and treatment of tumors (Yu et al., 2012b). Recent
the human promyelocytic leukemia cell line HL-6 and its subclone studies have suggested that miRNAs are involved in the regula-
cell line HP100, which is resistant to H2 O2 (Mizutani et al., 2005). tion of autophagy. As a member of the miRNAs family, miR-30a
Apoptosis of HL-6 but not HP100 cells was observed after cultur- has recently become a research hotspot in autophagy regulatio
ing the cells in the presence of doxorubicin for 7 h, suggesting that (Zhu et al., 2009). Our previous study demonstrated that miR-
H2 O2 is involved in doxorubicin-induced apoptosis in leukemia 30a inhibited autophagy by suppressing the expression of the
cells (Mizutani et al., 2005). Vincristine, a microtubule interfering autophagy-related gene Beclin-1. We observed increased expres-
anti-cancer agent, was found to be able to induce the production of sion of Beclin-1 and an increased number of LC3-positive autophagy
ROS in Jurkat cells at a very early stage, with the application of ROS events in cancer cells in which the expression of miR-30a was sup-
scavenger, Jurkat cells apoptosis was inhibited, which indicated pressed by cis-DDP or Taxol, suggesting that the chemotherapeutic
that ROS may play a key role in initiating apoptosis in Jurkat cells agents cis-DDP and Taxol improve autophagy of cancer cells by
(Groninger et al., 2002). Cytarabine, an important anti-metabolite inhibiting the expression of miR-30a (Zou et al., 2012). In addition,
used for the treatment of acute leukemia, can activate a number of lentiviral-induced over-expression of miR-30a down-regulated
tumor suppressor genes, such as NF-B and p53, by increasing the cancer cell autophagy in a Beclin-1-mediated manner (Zou et al.,
production of ROS, leading to apoptosis in leukemia cells (Romano 2012). These results suggest that miR-30a may be a potential
et al., 2000; Kanno et al., 2004). In addition, several natural ingre- target for resolving the drug resistance problem encountered in
dients and biological products, including parthenolide (Guzman cancer. Yu et al. found that miR-30a mimics or Beclin-1/ATG5
et al., 2005), Toona sinensis (Yang et al., 2006), gallic acid (Inoue knockout increased apoptosis in CML cells (Yu et al., 2012b).
et al., 1994), curcumin (Chen et al., 2005), resveratrol (Yaseen et al., In addition, antagomiR-30a promoted autophagy and inhibited
2012), cinnamaldehyde (Ka et al., 2003), and artesunate (Efferth imatinib-induced apoptosis in CML cells. These results suggest
et al., 2007), exhibit anti-leukemic effects by inducing leukemia that miR-30a inhibition is involved in the drug resistance of CML
cell apoptosis through ROS. cells (Yu et al., 2012b). A recent study demonstrated that p53, a
tumor suppressor protein, affected the chemotherapeutic effects of
leukemia by regulating leukemia cell autophagy. Cytoplasmic p53
6. Targeting autophagy to overcome drug resistance in inhibits autophagy; however, p53 in nuclei induces autophagy via
leukemia the expression of certain transcription factors (Feng et al., 2007).
It has been reported that p53 induces autophagy by activating the
Many studies have shown that a variety of drugs used to AMPK/mTOR pathway and upregulating damage-regulated mod-
treat leukemia cause autophagy. Wang et al. reported that Borte- ulator of AP (DRAM) (Feng et al., 2005). Amrein et al. found that
zomib inhibited proliferation, increased apoptosis, and activated p53-mediated autophagy was involved in dasatinib resistance in
autophagy in B-cell acute lymphoblastic leukemia (B-ALL) cells CLL cells (Amrein et al., 2011).
(Wang et al., 2015). However, resistance to Bortezomib was It is currently believed that autophagy is a double-edged sword
observed in B-ALL cells, and inhibitors of autophagy improved during the development of a tumor. Proper autophagy is impor-
the anti-leukemic activity of Bortezomib (Wang et al., 2015). tant for maintaining cell survival and cellular homeostasis, but
Evangelisti et al. also reported that triciribine induced apoptosis excessive autophagy can cause autophagic cell death, inhibiting
Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130 27

tumor growth (Gozuacik and Kimchi, 2007). Thus, the induction of resazurin (Erikstein et al., 2010), dihydroartemisinin (Wang et al.,
the autophagic death of tumor cells may be an effective approach 2012), and sesbania grandiora (Roy et al., 2013) have also been
to overcome the drug resistance of leukemia cells. Glucocorti- reported to induce autophagic cell death in leukemia cells through
coid (GC) is commonly used for the treatment of ALL; however, an increase in ROS production.
GC resistance is a common problem during the clinical treat-
ment of ALL. The activities of mTOR and AKT were found to be
increased in GC-resistant ALL cells, which inhibited autophagy in
these cells (Bornhauser et al., 2007). The combination of dexam- 8. Summary
ethasone and obatoclax, a Bcl-2 family antagonist, was found to
signicantly reduce the phosphorylation of mTOR-targeted pro- Autophagy is a common cellular activity that is involved in the
tein s6 and trigger autophagy-dependent programmed necrosis growth, differentiation, and proliferation of eukaryotic cells. ROS,
in ALL cells (Bonapace et al., 2010). Obatoclax also induces BAK- especially ROS produced in mitochondria, are signaling molecules
dependent apoptosis in ALL cells (Heidari et al., 2010). Given that that are involved in numerous signal transduction processes. The
obatoclax has a dual effect of inducing apoptosis and autophagic balance between ROS and autophagy plays an important role
cell death, the combination of obatoclax and dexamethasone may in maintaining cellular homeostasis. Both redox imbalance and
be able to overcome the drug resistance of ALL cells. It was reported mitophagy affect the homeostasis of cells and are involved in many
that the anti-leukemia effects of arsenic trioxide (As2 O3 ) in ALL also pathological processes. Numerous studies have shown that ROS and
depend mainly on the induction of autophagic cell death, and the autophagy play critically important roles in the development of
inhibition of autophagy blocks the anti-leukemia effects of As2 O3 leukemia. Current studies suggest that ROS and autophagy have
(Goussetis et al., 2010). The tyrosine kinase inhibitor imatinib is cur- extensive effects on cells and that the underlying mechanisms are
rently the rst-line drug for the treatment of CML. It was found that highly complicated. While a number of ROS-mediated autophagic
imatinib induced CML cell autophagy by increasing the expression pathways have been identied, details regarding the signaling
of Beclin-1 and ATG5, and it is believed that imatinib induced cell molecules and interactions among them are largely unknown.
death is achieved by autophagy. (Can et al., 2011). Taken together, In recent years, studies investigating ROS and autophagy have
these results suggest that the induction of leukemia cell autophagy signicantly improved our understanding of the pathogenesis of
through the use of drugs is an effective approach for the treatment leukemia and provide novel targets for the treatment of leukemia.
of leukemia. Many chemotherapeutic drugs that are currently used clinically for
the treatment of leukemia induce autophagic cell death and/or apo-
ptosis in leukemia cells by increasing ROS production. In addition,
7. ROS-induced autophagy as a therapeutic strategy for inhibition of ROS production represses the anti-leukemia effects
treatment of leukemia of chemotherapeutic drugs, suggesting that ROS play a critically
important role in the chemotherapy used to treat leukemia. How-
As an important signaling molecule that regulates cell ever, ROS are a double-edged sword in the treatment of tumors.
autophagy, ROS play an important role in autophagic cell death. High levels of ROS induce not only tumor cell death but also
Under normal conditions, ROS-induced autophagy reduces dam- oxidative damage to cell organelles, proteins, and DNA, leading to
age caused by oxidative stress to protect cells. However, ROS genomic instability and abnormality of cell function. In addition,
can also cause autophagic cell death under certain circumstances. ROS can activate NF-B, PI3K, MAPK, and other tumor growing
The mechanisms underlying the balance between ROS-induced signals. Low levels of ROS can improve the growth of tumor cells
autophagic protection and autophagic cell death remain unclear. rather than induce autophagy in tumor cells. Therefore, optimiza-
It has been reported that the activation of autophagy increases ROS tion of the concentration of intracellular ROS to achieve the best
production; however, caspase inhibition leads to the preferential anti-tumor effects is an important task. Monitoring the concentra-
degradation of catalase, resulting in the accumulation of ROS in tion of ROS in chemotherapy may allow physicians to better control
mitochondria and cell death. These results suggest that catalase the dose of chemotherapeutic drugs for individualized treatment.
degradation plays a critically important role in the balance between In addition, leukemia has a complex and diverse etiology, and dif-
ROS-induced autophagic protection and autophagic cell death (Yu ferent types of leukemia at different stages may be associated with
et al., 2006). distinct levels of ROS and autophagic activity. Therefore, ROS and
Numerous chemotherapeutic agents induce the autophagic cell autophagy have different roles in the development of leukemia,
death of tumor cells by increasing the production of ROS. Han and the same chemotherapeutic drugs may have different effects
et al. found that tetraarsenic hexoxide (As4 O6 ) inhibited the growth in different types and at different stages of leukemia.
and induced caspase-dependent apoptosis and Beclin-1-mediated Current knowledge regarding autophagy and autophagic cell
autophagic cell death in human leukemia U937 cells in a dose death is still limited. The signaling molecules and pathways
and time-dependent manner (Han et al., 2012). In addition, the involved in autophagy and its pathophysiological signicance
antioxidant N-acetylcysteine inhibited autophagic cell death and require further investigation. With an improved understanding of
apoptosis induced by As4 O6 , suggesting that ROS are involved in the molecular mechanisms underlying autophagy in the devel-
As4 O6 -induced autophagic cell death and apoptosis in U937 cells opment of tumors, especially the association between ROS and
(Han et al., 2012). Shinohara et al. reported that AIC-47, a fatty-acid autophagy, we may be able to identify more effective treatments
derivative, induced autophagic cell death in CML cells by down- for cancers by through the regulation of autophagy in tumor cells.
regulating the expression of C-Myc and the fused gene BCR-ABL
through the PPAR/-catenin pathway and increasing the pro-
duction of ROS (Shinohara et al., 2015). Eupalinin A, a natural
phytoalexin in Eupatorium chinense L., induces HL60 cell death by Conict of interest statement
increasing the production of ROS (Itoh et al., 2008). Eupalinin A has
been shown to increase the production of ROS and LC3II and to The authors have no other relevant afliations or nancial
reduce the mitochondrial membrane potential in HL60 cells, lead- involvement with any organizational entity with a nancial inter-
ing to HL60 cell death characterized mainly by autophagic cell death est in or nancial conicts with the subject matter or materials
(Itoh et al., 2008). In addition, piperlongumine (Xiong et al., 2015), discussed in the manuscript apart from those previously disclosed.
28 Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130

Funding Ekiz, H.A., Can, G., Baran, Y., 2012. Role of autophagy in the progression and
suppression of leukemias. Crit. Rev. Oncol. Hematol. 81, 275285.
Erikstein, B.S., Hagland, H.R., Nikolaisen, J., Kulawiec, M., Singh, K.K., Gjertsen, B.T.,
This study was supported by the Public Welfare Technology Tronstad, K.J., 2010. Cellular stress induced by resazurin leads to autophagy
Application Research Project of Zhejiang Province under Grant No. and cell death via production of reactive oxygen species and mitochondrial
2015C37122, Zhejiang, China, and the National Natural Science impairment. J. Cell. Biochem. 111, 574584.
Essick, E.E., Sam, F., 2010. Oxidative stress and autophagy in cardiac disease,
Foundation of China under Grant No. 81373139. neurological disorders, aging and cancer. Oxid. Med. Cell Longev. 3, 168177.
Evangelisti, C., Ricci, F., Tazzari, P., Chiarini, F., Battistelli, M., Falcieri, E., Ognibene,
A., Pagliaro, P., Cocco, L., McCubrey, J.A., et al., 2011. Preclinical testing of the
References Akt inhibitor triciribine in T-cell acute lymphoblastic leukemia. J. Cell. Physiol.
226, 822831.
Ahn, C.H., Jeong, E.G., Lee, J.W., Kim, M.S., Kim, S.H., Kim, S.S., Yoo, N.J., Lee, S.H., Evangelisti, C., Evangelisti, C., Chiarini, F., Lonetti, A., Buontempo, F., Neri, L.M.,
2007. Expression of beclin-1, an autophagy-related protein, in gastric and McCubrey, J.A., Martelli, A.M., 2015. Autophagy in acute leukemias: a
colorectal cancers. APMIS 115, 13441349. double-edged sword with important therapeutic implications. Biochim.
Altman, B.J., Jacobs, S.R., Mason, E.F., Michalek, R.D., MacIntyre, A.N., Coloff, J.L., Biophys. Acta 1853, 1426.
Ilkayeva, O., Jia, W., He, Y.W., Rathmell, J.C., 2011. Autophagy is essential to Farquhar, M.J., Bowen, D.T., 2003. Oxidative stress and the myelodysplastic
suppress cell stress and to allow BCR-Abl-mediated leukemogenesis. Oncogene syndromes. Int. J. Hematol. 77, 342350.
30, 18551867. Feng, Z., Zhang, H., Levine, A.J., Jin, S., 2005. The coordinate regulation of the p53
Amrein, L., Soulires, D., Johnston, J.B., 2011. Aloyz R. p53 and autophagy contribute and mTOR pathways in cells. Proc. Natl. Acad. Sci. U. S. A. 102, 82048209.
to dasatinib resistance in primary CLL lymphocytes. Leuk. Res. 35, 99102. Feng, Z., Hu, W., de Stanchina, E., Teresky, A.K., Jin, S., Lowe, S., Levine, A.J., 2007.
Aurelius, J., Thorn, F.B., Akhiani, A.A., Brune, M., Palmqvist, L., Hansson, M., The regulation of AMPK beta1, TSC2, and PTEN expression by p53: stress, cell
Hellstrand, K., Martner, A., 2012. Monocytic AML cells inactivate antileukemic and tissue specicity, and the role of these gene products in modulating the
lymphocytes: role of NADPH oxidase/gp91(phox) expression and the IGF-1-AKT-mTOR pathways. Cancer Res. 67, 30433053.
PARP-1/PAR pathway of apoptosis. Blood 119, 58325837. Frame, D., 2007. New strategies in controlling drug resistance in chronic myeloid
Battisti, V., Maders, L.D., Bagatini, M.D., Santos, K.F., Spanevello, R.M., Maldonado, leukemia. Am. J. Health. Syst. Pharm. 64, S1621.
P.A., Brul, A.O., Arajo Mdo, C., Schetinger, M.R., Morsch, V.M., 2008. Gobe, G., Crane, D., 2010. Mitochondria, reactive oxygen species and cadmium
Measurement of oxidative stress and antioxidant status in acute lymphoblastic toxicity in the kidney. Toxicol. Lett. 198, 4955.
leukemia patients. Clin. Biochem. 41 (78), 511518. Goussetis, D.J., Altman, J.K., Glaser, H., McNeer, J.L., Tallman, M.S., Platanias, L.C.,
Benhar, M., Dalyot, I., Engelberg, D., Levitzki, A., 2001. Enhanced ROS production in 2010. Autophagy is a critical mechanism for the induction of the antileukemic
oncogenically transformed cells potentiates c-Jun N-terminal kinase and p38 effects of arsenic trioxide. J. Biol. Chem. 285, 2998929997.
mitogen-activated protein kinase activation and sensitization to genotoxic Gozuacik, D., Kimchi, A., 2007. Autophagy and cell death. Curr. Top. Dev. Biol. 78,
stress. Mol. Cell. Biol. 21, 69136926. 217245.
Bonapace, L., Bornhauser, B.C., Schmitz, M., Cario, G., Ziegler, U., Niggli, F.K., Groninger, E., Meeuwsen-De Boer, G.J., De Graaf, S.S., Kamps, W.A., De Bont, E.S.,
Schfer, B.W., Schrappe, M., Stanulla, M., Bourquin, J.P., 2010. Induction of 2002. Vincristine induced apoptosis in acute lymphoblastic leukaemia cells: a
autophagy-dependent necroptosis is required for childhood acute mitochondrial controlled pathway regulated by reactive oxygen species? Int. J.
lymphoblastic leukemia cells to overcome glucocorticoid resistance. J. Clin. Oncol. 21, 13391345.
Invest. 120, 13101323. Guzman, M.L., Rossi, R.M., Karnischky, L., Li, X., Peterson, D.R., Howard, D.S., Jordan,
Bornhauser, B.C., Bonapace, L., Lindholm, D., Martinez, R., Cario, G., Schrappe, M., C.T., 2005. The sesquiterpene lactone parthenolide induces apoptosis of human
Niggli, F.K., Schfer, B.W., Bourquin, J.P., 2007. Low-dose arsenic trioxide acute myelogenous leukemia stem and progenitor cells. Blood 105, 41634169.
sensitizes glucocorticoid-resistant acute lymphoblastic leukemia cells to Han, M.H., Lee, W.S., Lu, J.N., Yun, J.W., Kim, G., Jung, J.M., Kim, G.Y., Lee, S.J., Kim,
dexamethasone via an Akt-dependent pathway. Blood 110, 20842091. W.J., Choi, Y.H., 2012. Tetraarsenic hexoxide induces beclin-1-induced
Boultwood, J., Fidler, C., Mills, K.I., Frodsham, P.M., Kusec, R., Gaiger, A., Gale, R.E., autophagic cell death as well as caspase-dependent apoptosis in U937 human
Linch, D.C., Littlewood, T.J., Moss, P.A., et al., 1996. Amplication of leukemic cells. Evid Based Compl. Altern. Med. 2012, 201414.
mitochondrial DNA in acute myeloid leukaemia. Br. J. Haematol. 95, 426431. He, C., Klionsky, D.J., 2009. Regulation mechanisms and signaling pathways of
Can, G., Ekiz, H.A., Baran, Y., 2011. Imatinib induces autophagy through BECLIN-1 autophagy. Annu. Rev. Genet. 43, 6793.
and ATG5 genes in chronic myeloid leukemia cells. Hematology 16, 9599. Heidari, N., Hicks, M.A., Harada, H., 2010. GX15-070 (obatoclax) overcomes
Cao, L., Xu, J., Lin, Y., Zhao, X., Liu, X., Chi, Z., 2009. Autophagy is upregulated in rats glucocorticoid resistance in acute lymphoblastic leukemia through induction
with status epilepticus and partly inhibited by Vitamin E. Biochem. Biophys. of apoptosis and autophagy. Cell. Death. Dis. 1, e76.
Res. Commun. 379, 949953. Herrera-Goepfert, R., Barrios-Del Valle, R., Sales-Carmona, V., Santoyo, J.,
Changchien, J.J., Chen, Y.J., Huang, C.H., Cheng, T.L., Lin, S.R., Chang, L.S., 2015. Oliva-Ramirez, E.B., 1986. Intramitochondrial lamellar bodies in acute
Quinacrine induces apoptosis in human leukemia K562 cells via p38 myeloblastic leukemia. Hum. Pathol. 17, 748753.
MAPK-elicited BCL2 down-regulation and suppression of ERK/c-Jun-mediated Hole, P.S., Zabkiewicz, J., Munje, C., Newton, Z., Pearn, L., White, P., Marquez, N.,
BCL2L1 expression. Toxicol. Appl. Pharmacol. 284, 3341. Hills, R.K., Burnett, A.K., Tonks, A., Darley, R.L., 2013. Overproduction of
Chauncey, T.R., 2001. Drug resistance mechanisms in acute leukemia. Curr. Opin. NOX-derived ROS in AML promotes proliferation and is associated with
Oncol. 13, 2126. defective oxidativestress signaling. Blood 122 (19), 33223330.
Chen, J., Wanming, D., Zhang, D., Liu, Q., Kang, J., 2005. Water-soluble antioxidants Houwerzijl, E.J., Pol, H.W., Blom, N.R., van der Want, J.J., de Wolf, J.T., Vellenga, E.,
improve the antioxidant and anticancer activity of low concentrations of 2009. Erythroid precursors from patients with low-risk myelodysplasia
curcumin in human leukemia cells. Pharmazie 60, 5761. demonstrate ultrastructural features of enhanced autophagy of mitochondria.
Chen, P., Cescon, M., Bonaldo, P., 2014a. Autophagy-mediated regulation of Leukemia 23, 886891.
macrophages and its applications for cancer. Autophagy 10, 192200. Hu, B., Yue, Q.F., Chen, Y., Bu, F.D., Sun, C.Y., Liu, X.Y., 2015. Expression of autophagy
Chen, Y., Luo, G., Yuan, J., Wang, Y., Yang, X., Wang, X., Li, G., Liu, Z., Zhong, N., related gene BECLIN-1 and number of autophagic vacuoles in bone marrow
2014b. Vitamin C mitigates oxidative stress and tumor necrosis factor-alpha in mononuclear cells from 40 myelodysplastic syndromes patients and their
severe community-acquired pneumonia and LPS-induced macrophages. signicance. Zhongguo Shi Yan Xue Ye Xue Za Zhi 23, 146149.
Mediators Inamm. 2014, 426740. Hwang, I.T., Chung, Y.M., Kim, J.J., Chung, J.S., Kim, B.S., Kim, H.J., Kim, J.S., Yoo, Y.D.,
Chiaretti, S., Gianfelici, V., Ceglie, G., Fo, R., 2014. Genomic characterization of 2007. Drug resistance to 5-FU linked to reactive oxygen species modulator 1.
acute leukemias. Med. Princ. Pract. 23, 487506. Biochem. Biophys. Res. Commun. 359, 304310.
Clarke, P.G., Puyal, J., 2012. Autophagic cell death exists. Autophagy 8, 867869. Inoue, M., Suzuki, R., Koide, T., Sakaguchi, N., Ogihara, Y., Yabu, Y., 1994.
Collado, R., Oliver, I., Tormos, C., Egea, M., Miguel, A., Cerd, C., Ivars, D., Borrego, S., Antioxidant, gallic acid, induces apoptosis in HL-60RG cells. Biochem. Biophys.
Carbonell, F., Sez, G.T., 2012. Early ROS-mediated DNA damage and oxidative Res. Commun. 204, 898904.
stress biomarkers in monoclonal B lymphocytosis. Cancer Lett. 317, 144149. Itoh, T., Ito, Y., Ohguchi, K., Ohyama, M., Iinuma, M., Otsuki, Y., Nozawa, Y., Akao, Y.,
Dadakhujaev, S., Jung, E.J., Noh, H.S., Hah, Y.S., Kim, C.J., Kim, D.R., 2009. Interplay 2008. Eupalinin: an isolated from Eupatorium chinense L. induces
between autophagy and apoptosis in TrkA-induced cell death. Autophagy 5, autophagocytosis in human leukemia HL60 cells. Bioorg. Med. Chem. 16,
103105. 721731.
Devi, G.S., Prasad, M.H., Saraswathi, I., Raghu, D., Rao, D.N., Reddy, P.P., 2000. Free Iwama, Y., Eguchi, M., 1986. Quantitative evaluation of leukemic mitochondria
radicals antioxidant enzymes and lipid peroxidation in different types of with a computer-controlled image analyzer. Virchows Arch. B. Cell Pathol. Incl.
leukemias. Clin. Chim. Acta 293 (12), 5362. Mol. Pathol. 51, 375384.
Dickinson, B.C., Chang, C.J., 2011. Chemistry and biology of reactive oxygen species Jang, Y.Y., Sharkis, S.J., 2007. A low level of reactive oxygen species selects for
in signaling or stress responses. Nat. Chem. Biol. 7 (8), 504511. primitive hematopoietic stem cells that may reside in the low-oxygenic niche.
Dong, J.M., Zhao, S.G., Huang, G.Y., Liu, Q., 2004. NADPH oxidase-mediated Blood 10, 30563063.
generation of reactive oxygen species is critically required for survival of Jung, H.S., Lee, M.S., 2010. Role of autophagy in diabetes and mitochondria. Ann. N.
undifferentiated human promyelocytic leukemia cell line HL-60. Free Radic. Y. Acad. Sci. 1201, 7983.
Res. 38 (6), 629637. Juntilla, M.M., Patil, V.D., Calamito, M., Joshi, R.P., Birnbaum, M.J., Koretzky, G.A.,
Efferth, T., Giaisi, M., Merling, A., Krammer, P.H., Li-Weber, M., 2007. Artesunate 2010. AKT1 and AKT2 maintain hematopoietic stem cell function by regulating
induces ROS-mediated apoptosis in doxorubicin-resistant T leukemia cells. reactive oxygen species. Blood 115, 40304038.
PLoS One 2, e693.
Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130 29

Ka, H., Park, H.J., Jung, H.J., Choi, J.W., Cho, K.S., Ha, J., Lee, K.T., 2003. syndrome, fanconi anaemia and werner syndrome. Biogerontology 11,
Cinnamaldehyde induces apoptosis by ROS-mediated mitochondrial 401419.
permeability transition in human promyelocytic leukemia HL-60 cells. Cancer Popp, H.D., Bohlander, S.K., 2010. Genetic instability in inherited and sporadic
Lett. 196, 143152. leukemias. Genes. Chromosomes Cancer 49 (12), 10711081.
Kanno, S., Higurashi, A., Watanabe, Y., Shouji, A., Asou, K., Ishikawa, M., 2004. Rassool, F.V., Gaymes, T.J., Omidvar, N., Brady, N., Beurlet, S., Pla, M., Reboul, M.,
Susceptibility to cytosine arabinoside (Ara-C)-induced cytotoxicity in human Lea, N., Chomienne, C., Thomas, N.S., et al., 2007. Reactive oxygen species, DNA
leukemia cell lines. Toxicol. Lett. 152, 149158. damage, and error-prone repair: a model for genomic instability with
Karantza-Wadsworth, V., White, E., 2007. Role of autophagy in breast cancer. progression in myeloid leukemia. Cancer Res. 67, 87628771.
Autophagy 3, 610613. Richardson, C., Yan, S., Vestal, C.G., 2015. Oxidative stress, bone marrow failure, and
Kim, I.G., Kim, J.S., Lee, J.H., Cho, E.W., 2014. Genistein decreases cellular redox genome instability in hematopoietic stem cells. Int. J. Mol. Sci. 16, 23662385.
potential, partially suppresses cell growth in HL-60 leukemia cells and Romano, M.F., Lamberti, A., Bisogni, R., Tassone, P., Pagnini, D., Storti, G., Del
sensitizes cells to -radiation-induced cell death. Mol. Med. Rep. 10, Vecchio, L., Turco, M.C., Venuta, S., 2000. Enhancement of cytosine
27862792. arabinoside-induced apoptosis in human myeloblastic leukemia cells by
Kobashigawa, S., Kashino, G., Suzuki, K., Yamashita, S., Mori, H., 2015. Ionizing NF-kappa B/Rel- specic decoy oligodeoxynucleotides. Gene Ther. 7,
radiation-induced cell death is partly caused by increase of mitochondrial 12341237.
reactive oxygen species in normal human broblast cells. Radiat. Res. 183, Roy, R., Kumar, D., Chakraborty, B., Chowdhury, C., Das, P., 2013. Apoptotic and
455464. autophagic effects of Sesbania grandiora owers in human leukemic cells.
Kongsuphol, P., Mukda, S., Nopparat, C., Villarroel, A., Govitrapong, P., 2009. PLoS One 8, e71672.
Melatonin attenuates methamphetamine-induced deactivation of the Ruvolo, V.R., Karanjeet, K.B., Schuster, T.F., Brown, R., Deng, Y., Hinchcliffe, E.,
mammalian target of rapamycin signaling to induce autophagy in SK-N-SH Ruvolo, P.P., 2010. Role for PKC in fenretinide-mediated apoptosis in
cells. J. Pineal Res. 46, 199206. lymphoid leukemia cells. J. Signal. Transduct. 2010, 584657.
Kwak, H.J., Liu, P., Bajrami, B., Xu, Y., Park, S.Y., Nombela-Arrieta, C., Mondal, S., Sun, Salemi, S., Youse, S., Constantinescu, M.A., Fey, M.F., Simon, H.U., 2012. Autophagy
Y., Zhu, H., Chai, L., et al., 2015. Myeloid cell-derived reactive oxygen species is required for self-renewal and differentiation of adult human stem cells. Cell
externally regulate the proliferation of myeloid progenitors in emergency Res. 22, 432435.
granulopoiesis. Immunity 42, 159171. Samanta, A., Perazzona, B., Chakraborty, S., Sun, X., Modi, H., Bhatia, R., Priebe, W.,
Landry, W.D., Cotter, T.G., 2014. ROS signalling, NADPH oxidases and cancer. Arlinghaus, R., 2011. Janus kinase 2 regulates Bcr-Abl signaling in chronic
Biochem. Soc. Trans. 42 (4), 934938. myeloid leukemia. Leukemia 25, 463472.
Lee, W.L., Huang, J.Y., Shyur, L.F., 2013. Phytoagents for cancer management: Sattler, M., Verma, S., Shrikhande, G., Byrne, C.H., Pride, Y.B., Winkler, T.,
regulation of nucleic acid oxidation, ROS, and related mechanisms. Oxid. Med. Greeneld, E.A., Salgia, R., Grifn, J.D., 2000. The BCR/ABL tyrosine kinase
Cell Longev. 2013, 925804. induces production of reactive oxygen species in hematopoietic cells. J. Biol.
Lewandowski, D., Barroca, V., Ducong, F., Bayer, J., Van Nhieu, J.T., Pestourie, C., Chem. 275, 2427324278.
Fouchet, P., Tavitian, B., Romo, P.H., 2010. In vivo cellular imaging pinpoints Schumacher, H.R., Szekely, I.E., Fisher, D.R., 1975. Leukemic mitochondria. III. Acute
the role of reactive oxygen species in the early steps of adult hematopoietic lymphoblastic leukemia. Am. J. Pathol. 78, 4958.
reconstitution. Blood 115, 443452. Shao, L., Li, H., Pazhanisamy, S.K., Meng, A., Wang, Y., Zhou, D., 2011. Reactive
Li, L., Tan, J., Miao, Y., Lei, P., Zhang, Q., 2015. ROS and autophagy: interactions and oxygen species and hematopoietic stem cell senescence. Int. J. Hematol. 94,
molecular regulatory mechanisms. Cell. Mol. Neurobiol. 35, 615621. 2432.
Libik-Konieczny, M., Kozieradzka-Kiszkurno, M., Desel, C., Michalec-Warzecha, Z., She, M.R., Li, J.G., Guo, K.Y., Lin, W., Du, X., Niu, X.Q., 2007. Requirement of reactive
Miszalski, Z., Konieczny, R., 2015. The localization of NADPH oxidase and oxygen species generation in apoptosis of leukemia cells induced by
reactive oxygen species in vitro-cultured Mesembryanthemum crystallinum L. 2-methoxyestradiol. Acta Pharmacol. Sin. 28, 10371044.
hypocotyls discloses their differing roles in rhizogenesis. Protoplasma 252 (2), Shinohara, H., Taniguchi, K., Kumazaki, M., Yamada, N., Ito, Y., Otsuki, Y., Uno, B.,
477487. Hayakawa, F., Minami, Y., Naoe, T., et al., 2015. Anti-cancer fatty-acid
Lin, X., Fang, Q., Chen, S., Zhe, N., Chai, Q., Yu, M., Zhang, Y., Wang, Z., Wang, J., derivative induces autophagic cell death through modulation of PKM isoform
2015. Heme oxygenase-1 suppresses the apoptosis of acute myeloid leukemia expression prole mediated by bcr-abl in chronic myeloid leukemia. Cancer
cells via the JNK/c-JUN signaling pathway. Leuk. Res. 39 (5), 544552. Lett. 360, 2838.
Lisanti, M.P., Martinez-Outschoorn, U.E., Chiavarina, B., Pavlides, S., Silber, R., Farber, C.M., Papadopoulos, E., Nevrla, D., Liebes, L., Bruck, M., Brown, R.,
Whitaker-Menezes, D., Tsirigos, A., Witkiewicz, A., Lin, Z., Balliet, R., Howell, A., Canellakis, Z.N., 1992. Glutathione depletion in chronic lymphocytic leukemia
et al., 2010. Understanding the lethal drivers of tumor-stroma co-evolution: B lymphocytes. Blood 80 (8), 20382043.
emerging role(s) for hypoxia, oxidative stress and autophagy/mitophagy in the Singh, M.M., Irwin, M.E., Gao, Y., Ban, K., Shi, P., Arlinghaus, R.B., Amin, H.M.,
tumor micro-environment. Cancer Biol. Ther. 10, 537542. Chandra, J., 2012. Inhibition of the NADPH oxidase regulates heme oxygenase 1
Liu, L., Yang, M., Kang, R., Wang, Z., Zhao, Y., Yu, Y., Xie, M., Yin, X., Livesey, K.M., expression in chronic myeloid leukemia. Cancer 118 (13), 34333445.
Lotze, M.T., et al., 2011. HMGB1-induced autophagy promotes chemotherapy Stankov, M.V., El Khatib, M., Kumar Thakur, B., Heitmann, K.,
resistance in leukemia cells. Leukemia 25, 2331. Panayotova-Dimitrova, D., Schoening, J., Bourquin, J.P., Schweitzer, N.,
Ludin, A., Gur-Cohen, S., Golan, K., Kaufmann, K.B., Itkin, T., Medaglia, C., Lu, X.J., Leverkus, M., Welte, K., et al., 2014. Histone deacetylase inhibitors induce
Ledergor, G., Kollet, O., Lapidot, T., 2014. Reactive oxygen species regulate apoptosis in myeloid leukemia by suppressing autophagy. Leukemia 28,
hematopoietic stem cell self-renewal, migration and development, as well as 577588.
their bone marrow microenvironment. Antioxid. Redox Signal. 21, 16051619. Stella, S., Tirr, E., Conte, E., Stagno, F., Di Raimondo, F., Manzella, L., Vigneri, P.,
Lyu, B.N., Ismailov, S.B., Ismailov, B., Lyu, M.B., 2008. Mitochondrial concept of 2013. Suppression of survivin induced by a BCR-ABL/JAK2/STAT3 pathway
leukemogenesis: key role of oxygen-peroxide effects. Theor. Biol. Med. Model. sensitizes imatinib-resistant CML cells to different cytotoxic drugs. Mol.
5, 23. Cancer Ther. 12, 10851098.
Mizutani, H., Tada-Oikawa, S., Hiraku, Y., Kojima, M., Kawanishi, S., 2005. Sun, Y., Liu, J.H., Jin, L., Lin, S.M., Yang, Y., Sui, Y.X., Shi, H., 2010. Over-expression of
Mechanism of apoptosis induced by doxorubicin through the generation of the Beclin1 gene upregulates chemosensitivity to anti-cancer drugs by
hydrogen peroxide. Life Sci. 76, 14391453. enhancing therapy-induced apoptosis in cervix squamous carcinoma CaSki
Montero, A.J., Jassem, J., 2011. Cellular redox pathways as a therapeutic target in cells. Cancer Lett. 294, 204210.
the t reatment of cancer. Drugs 71, 13851396. Szekely, I.E., Fischer, D.R., Schumacher, H.R., 1976. Leukemic mitochondria. II.
Mortensen, M., Soilleux, E.J., Djordjevic, G., Tripp, R., Lutteropp, M., Sadighi-Akha, Acute monoblastic leukemia. Cancer 37, 805811.
E., Stranks, A.J., Glanville, J., Knight, S., Jacobsen, S.E., et al., 2011. The Tang, H., Da, L., Mao, Y., Li, Y., Li, D., Xu, Z., Li, F., Wang, Y., Tiollais, P., Li, T., et al.,
autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J. 2009. Hepatitis B virus X protein sensitizes cells to starvation-induced
Exp. Med. 208, 455467. autophagy via up-regulation of beclin 1 expression. Hepatology 49, 6071.
Naka, K., Muraguchi, T., Hoshii, T., Hirao, A., 2008. Regulation of reactive oxygen Tien Kuo, M., Savaraj, N., 2006. Roles of reactive oxygen species in
species and genomic stability in hematopoietic stem cells. Antioxid. Redox hepatocarcinogenesis and drug resistance gene expression in liver cancers.
Signal. 10, 18831894. Mol. Carcinog. 45, 701709.
Nogueira, V., Hay, N., 2013a. Molecular pathways: reactive oxygen species Tonino, S.H., van Laar, J., van Oers, M.H., Wang, J.Y., Eldering, E., Kater, A.P., 2011.
homeostasis in cancer cells and implications for cancer therapy. Clin. Cancer ROS-mediated upregulation of Noxa overcomes chemoresistance in chronic
Res. 19, 43094314. lymphocytic leukemia. Oncogene 30, 701713.
Nogueira, V., Hay, N., 2013b. Molecular pathways: reactive oxygen species Torgersen, M.L., Simonsen, A., 2013. Autophagy: friend or foe in the treatment of
homeostasis in cancer cells and implications for cancer therapy. Clin. Cancer fusion protein-associated leukemias. Autophagy 9, 21752177.
Res. 19, 43094314. Udensi, U.K., Tchounwou, P.B., 2014. Dual effect of oxidative stress on leukemia
Oltra, A.M., Carbonell, F., Tormos, C., Iradi, A., Sez, G.T., 2001. Antioxidant enzyme cancer induction and treatment. J. Exp. Clin. Cancer Res. 33, 106.
activities and the production of MDA and 8-oxo-dG in chronic lymphocytic Underwood, B.R., Imarisio, S., Fleming, A., Rose, C., Krishna, G., Heard, P., Quick, M.,
leukemia. Free Radic. Biol. Med. 30 (11), 12861292. Korolchuk, V.I., Renna, M., Sarkar, S., et al., 2010. Antioxidants can inhibit basal
Owusu-Ansah, E., Banerjee, U., 2009. Reactive oxygen species prime Drosophila autophagy and enhance neurodegeneration in models of polyglutamine
haematopoietic progenitors for differentiation. Nature 461, 537541. disease. Hum. Mol. Genet. 19, 34133429.
Pallard, F.V., Lloret, A., Lebel, M., dIschia, M., Cogger, V.C., Le Couteur, D.G., Wan, S.Y., Zhang, R., Wang, Y.Y., Cen, J.N., Zhou, J., Yang, Y., Jiang, F., Chen, Z.X., 2013.
Gadaleta, M.N., Castello, G., Pagano, G., 2010. Mitochondrial dysfunction in Expression of autophagy related gene Beclin1 in myelodysplastic syndrome
some oxidative stress-related genetic diseases: ataxia-telangiectasia, down patients and its signicance. Zhongguo Shi Yan Xue Ye Xue Za Zhi 21, 936939.
30 Y.-F. Chen et al. / Critical Reviews in Oncology/Hematology 112 (2017) 2130

Wang, Z., Hu, W., Zhang, J.L., Wu, X.H., Zhou, H.J., 2012. Dihydroartemisinin induces Yorimitsu, T., Klionsky, D.J., 2007. Eating the endoplasmic reticulum: quality
autophagy and inhibits the growth of iron-loaded human myeloid leukemia control by autophagy. Trends Cell Biol. 17, 279285.
K562 cells via ROS toxicity. FEBS Open Biol. 2, 103112. Yu, L., Wan, F., Dutta, S., Welsh, S., Liu, Z., Freundt, E., Baehrecke, E.H., Lenardo, M.,
Wang, Z., Zhu, S., Zhang, G., Liu, S., 2015. Inhibition of autophagy enhances the 2006. Autophagic programmed cell death by selective catalase degradation.
anticancer activity of bortezomib in B-cell acute lymphoblastic leukemia cells. Proc. Natl. Acad. Sci. U. S. A. 103, 49524957.
Am. J. Cancer. Res. 5, 639650. Yu, Y., Xie, M., Kang, R., Livesey, K.M., Cao, L., Tang, D., 2012a. HMGB1 is a
Watson, A.S., Mortensen, M., Simon, A.K., 2011a. Autophagy in the pathogenesis of therapeutic target for leukemia. Am. J. Blood Res. 2, 3643.
myelodysplastic syndrome and acute myeloid leukemia. ABBV Cell Cycle 10, Yu, Y., Cao, L., Yang, L., Kang, R., Lotze, M., Tang, D., 2012b. MicroRNA 30A promotes
17191725. autophagy in response to cancer therapy. Autophagy 8, 853855.
Watson, A.S., Mortensen, M., Simon, A.K., 2011b. Autophagy in the pathogenesis of Zare-Abdollahi, D., Safari, S., Movafagh, A., Ghadiani, M., Tabarraee, M.,
myelodysplastic syndrome and acute myeloid leukemia. ABBV Cell Cycle 10, Riazi-Isfahani, S., Gorji, S., Keyvan, L., Gachkar, L., 2016. Expression analysis of
17191725. BECN1 in acute myeloid leukemia: association with distinct cytogenetic and
Xiong, X.X., Liu, J.M., Qiu, X.Y., Pan, F., Yu, S.B., Chen, X.Q., 2015. Piperlongumine molecular abnormalities. Int. J. Lab. Hematol. 38, 125132.
induces apoptotic and autophagic death of the primary myeloid leukemia cells Zhou, Y., Hileman, E.O., Plunkett, W., Keating, M.J., Huang, P., 2003. Free radical
from patients via activation of ROS-p38/JNK pathways. Acta Pharmacol. Sin. 36, stress in chronic lymphocytic leukemia cells and its role in cellular sensitivity
362374. to ROS-generating anticancer agents. Blood 101, 40984104.
Xu, Y., Kim, S.O., Li, Y., Han, J., 2006. Autophagy contributes to Zhou, F.L., Zhang, W.G., Wei, Y.C., Meng, S., Bai, G.G., Wang, B.Y., Yang, H.Y., Tian,
caspase-independent macrophage cell death. J. Biol. Chem. 281, 1917919187. W., Meng, X., Zhang, H., et al., 2010. Involvement of oxidative stress in the
Yahata, T., Takanashi, T., Muguruma, Y., Ibrahim, A.A., Matsuzawa, H., Uno, T., relapse of acute myeloid leukemia. J. Biol. Chem. 285, 1501015015.
Sheng, Y., Onizuka, M., Ito, M., Kato, S., et al., 2011. Accumulation of oxidative Zhou, F., Shen, Q., Claret, F.X., 2013. Novel roles of reactive oxygen species in the
DNA damage restricts the self-renewal capacity of human hematopoietic stem pathogenesis of acute myeloid leukemia. J. Leukoc. Biol. 94, 423429.
cells. Blood 118, 29412950. Zhu, H., Wu, H., Liu, X., Li, B., Chen, Y., Ren, X., Liu, C.G., Yang, J.M., 2009. Regulation
Yang, H.L., Chang, W.H., Chia, Y.C., Huang, C.J., Lu, F.J., Hsu, H.K., Hseu, Y.C., 2006. of autophagy by a beclin 1-targeted microRNA, miR-30a, in cancer cells.
Toona sinensis extracts induces apoptosis via reactive oxygen species in Autophagy 5, 816823.
human premyelocytic leukemia cells. Food Chem. Toxicol. 44, 19781988. Zou, Z., Wu, L., Ding, H., Wang, Y., Zhang, Y., Chen, X., Chen, X., Zhang, C.Y., Zhang,
Yaseen, A., Chen, S., Hock, S., Rosato, R., Dent, P., Dai, Y., Grant, S., 2012. Resveratrol Q., Zen, K., 2012. MicroRNA-30a sensitizes tumor cells to cis-platinum via
sensitizes acute myelogenous leukemia cells to histone deacetylase inhibitors suppressing beclin 1-mediated autophagy. J. Biol. Chem. 287, 41484156.
through reactive oxygen species-mediated activation of the extrinsic apoptotic de Moura, M.B., dos Santos, L.S., Van Houten, B., 2010. Mitochondrial dysfunction in
pathway. Mol. Pharmacol. 82, 10301041. neurodegenerative diseases and cancer. Environ. Mol. Mutagen. 51, 391405.
Yi, J., Gao, F., Shi, G., Li, H., Wang, Z., Shi, X., Tang, X., 2002. The inherent cellular
level of reactive oxygen species: one of the mechanisms determining apoptotic
susceptibility of leukemic cells to arsenic trioxide. Apoptosis 7, 209215.

S-ar putea să vă placă și