You are on page 1of 11

Chemico-Biological Interactions 284 (2018) 1–11

Contents lists available at ScienceDirect

Chemico-Biological Interactions
journal homepage: www.elsevier.com/locate/chembioint

Drug-induced thrombocytopenia: Focus on platelet apoptosis T


a,b a,b,c,∗
Enoli De Silva , Hugh Kim
a
Centre for Blood Research, University of British Columbia, Vancouver, Canada
b
Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
c
Faculty of Dentistry, University of British Columbia, Vancouver, Canada

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

Keywords: Thrombocytopenia is a serious and potentially fatal complication of drug therapy that results either from a
Drugs decrease in bone marrow platelet production or the excessive destruction of circulating platelets. Although
Thrombocytopenia multiple mechanisms are responsible for deregulated platelet clearance, the role of programmed platelet death
Platelets (apoptosis) in drug-induced thrombocytopenia has been relatively under-investigated until recently. Here we
Apoptosis
review apoptotic signaling pathways in platelets, with a focus on current data that provide mechanistic insights
into drug-induced apoptosis and thrombocytopenia.

1. Introduction configurations of the drug and antibody molecules interacting at the


platelet surface. These subtypes of immune-mediated platelet destruc-
Platelets are anucleate blood cells essential for hemostasis and tion are summarized in Table 1.
wound healing; tight regulation of platelet numbers is crucial for
human health. Platelets are synthesized and released from bone marrow 3. Nonimmune-mediated platelet destruction
megakaryocytes into the circulation where they remain for 7–10 days
[1]. In humans, the normal circulating platelet count ranges from In contrast to immune-mediated DIT, nonimmune-mediated platelet
150,000–450,000 platelets/μl of whole blood. A reduction in platelet destruction is described as a direct cytotoxic effect of the drug mole-
count below 150,000 platelets/μl is termed thrombocytopenia, a con- cules on the platelets. For example, thrombocytopenia was previously
dition that increases the risk of potentially life-threatening hemorrhage reported to result from the chemotherapeutic administration of inter-
[2]. Drug-induced thrombocytopenia (DIT) can occur following ad- feron-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). The
ministration of a wide range of medications, including antibiotics, purported mechanism was attributed to altered platelet-endothelial cell
cardiac drugs and anti-neoplastic agents [3]. DIT-inducing agents can interactions, and therefore, antibody-independent [8,9]. However, over
perturb platelet counts in one of 2 ways: centrally, by exerting cytotoxic the past 7 years, considerable evidence has emerged in support of an-
effects on bone marrow megakaryocytes, thus reducing platelet synth- other form of nonimmune-mediated platelet destruction: platelet
esis (Fig. 1A); or peripherally, by enhancing clearance of platelets al- apoptosis, or programmed cell death. Published evidence from multiple
ready in circulation (Fig. 1B). Drugs can accelerate the destruction of groups clearly indicate that thrombocytopenia-inducing drugs at-
circulating platelets through both immune- and nonimmune-mediated tenuate platelet numbers by triggering pro-apoptotic signaling.
mechanisms.
4. Pro-apoptotic signaling in platelets
2. Immune-mediated platelet destruction
Apoptosis is a programmed form of cell death that results in a
In immune-mediated platelet destruction, drug-dependent anti- controlled clearance of cells by macrophages, without eliciting an in-
bodies bind to platelets, either directly or alongside accessory proteins flammatory response. Apoptosis is essential for the removal of damaged
[4]. Platelets are then phagocytosed by macrophages that recognize the cells from the body and maintaining appropriate cell populations in
drug-dependent antibody on the platelet surface. The subject of im- developing tissues [10]. This form of cell death contrasts with necrosis,
mune-mediated DIT is covered in-depth by several excellent review which is characterized by the uncontrolled rupture and release of cel-
articles [3,5–7] and as such is only briefly mentioned here. There are 6 lular contents [11], resulting in tissue damage. The recognition that
subtypes of immune-mediated DIT that reflect different spatial platelets undergo apoptosis is a relatively new concept that has been


Corresponding author. UBC Centre for Blood Research, 2350 Health Sciences Mall, 4th Floor, Vancouver, BC, V6T 1Z3, Canada.
E-mail address: hughkim@dentistry.ubc.ca (H. Kim).

https://doi.org/10.1016/j.cbi.2018.01.015
Received 31 July 2017; Received in revised form 23 December 2017; Accepted 18 January 2018
Available online 02 February 2018
0009-2797/ © 2018 Elsevier B.V. All rights reserved.
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

Fig. 1. Platelet production and clearance. A. Decreased platelet production by megakaryocytes (MK) in the bone marrow can result from impaired megakaryopoiesis, thrombopoietin
(TPO) signaling defects and drug-induced myelosuppression. B. Circulating platelets may be prematurely cleared due to sequestration in the spleen, autoantibodies against platelet
glycoproteins and drug-induced platelet destruction via both immune- and non-immune mediated mechanisms. De-regulation of platelet production and/or clearance can lead to
thrombocytopenia.

Table 1
Mechanisms of immune-mediated platelet destruction.

Name Mechanism Typical drug Refs

Hapten-induced antibody • Drug (hapten) covalently binds to the amine groups of proteins, which elicits an immune response Antibiotics [3,5]
Drug-dependent antibody • Drug associates with platelet glycoproteins that is then targeted by antibodies Quinine [3,5]
• Drug exposes glycoprotein sites that are readily recognized by the immune system
• Immune response requires a soluble drug
Drug-induced autoantibodies • Autoantibody directly targets platelets Procainamide [5,9]
• Drug exposes glycoprotein sites that are readily recognized by the immune system, which triggers production
of autoantibodies
Immune complex • complex
Heparin binds and structurally modifies platelet factor 4 causing the formation of antibodies against this Heparin [3,5,97]

Drug-specific antibody • Aoccurring


chimeric (human/mouse) Fab domain that binds βA domain of GPIIIa, which is recognized by a naturally
antibody or drug-sensitive antibody
Abciximab [3,5,97]

Fiban-induced thrombocytopenia • Drug binds to the arginine-glycine-aspartic acid (RGD) recognition site on platelet GPIIb/IIIa receptor, which is
recognized by a naturally occurring antibody
Tirofiban [3,5]

increasingly documented over the past 10–15 years [12–14]. signaling cascade leading to caspase-8 activation, followed by caspase-3
As is observed in nucleated cells, apoptotic platelets typically ex- activation and apoptosis [10]. There is relatively little evidence to
hibit cytoplasmic shrinkage, membrane protrusions known as “blebs” suggest that platelets undergo extrinsic apoptosis as they do not express
and increased cell surface exposure of phosphatidylserine (PS) [10,15] the prototypical Fas death receptor [13]. However, this contention is
which denotes a cell's slated clearance by phagocytes [16]. Pro-apop- mitigated by the finding that platelets do express caspase-8, a marker of
totic signaling pathways are classified as extrinsic or intrinsic (Fig. 2). extrinsic apoptosis [17]. Moreover, thrombin, a platelet agonist and
Extrinsic apoptosis is specifically mediated by extracellular ligand ligand for the protease-activated receptor-1 (PAR-1) [18], reportedly
binding to “death” receptors, however, both pathways culminate in the induces caspase-3 activation and cell death [19]. This suggests the ex-
activation of caspases 3 and 7, cysteine proteases that orchestrate cell istence in platelets of an extrinsic apoptotic pathway that operates in-
death by degrading cellular proteins. Therefore, the de-regulation of dependently of the classical death receptors. Consequently, the precise
normal apoptotic signaling mechanisms would plausibly explain am- determinants of receptor-mediated platelet apoptosis remain undefined
plified platelet destruction observed in drug-induced thrombocyto- at this time.
penia.

4.2. Intrinsic apoptosis


4.1. Extrinsic (receptor-mediated) apoptosis
In contrast to the extrinsic apoptosis pathway, intrinsic apoptosis is
The extrinsic apoptosis pathway is initiated by the binding of ex- a receptor-independent process initiated by stimuli such as radiation,
tracellular ligands with cell surface receptors. For example, members of toxins, free radicals or an increase in intracellular calcium concentra-
the tumor necrosis factor (TNF) receptor superfamily (FasR, TNFR1, tion ([Ca2+]i) [10]. A central feature of intrinsic apoptosis is depolar-
DR3, DR4 and DR5) are activated by their respective ligands (FasL, ization of the mitochondria, and this is regulated by several pro-apop-
TNF-α, Apo3L, Apo2L, Apo2L) [10]. Death receptor ligation triggers a totic and anti-apoptotic accessory proteins (Fig. 2).

2
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

Fig. 2. The intrinsic apoptosis pathway. Schematic diagram depicts the steps in the intrinsic apoptosis pathway. BH3-only domain pro-death proteins are induced by cellular stress and/or
pro-apoptotic chemical stimuli (1). In the resting cell, multi-domain pro-survival proteins suppress activation of multi-domain pro-apoptotic proteins, Bax/Bak (depicted in red).
Activated BH3-only domain pro-death proteins can activate Bax/Bak via two mechanisms: by inhibiting Bcl-2 pro-survival proteins thereby removing their suppression of Bax/Bak (2a);
or by directly activating Bax/Bak (2b). Activated Bax translocates to the mitochondria and oligomerizes on the mitochondrial membrane (3). Bax/Bak mediate mitochondrial outer
membrane permeabilization (MOMP) (4). The loss of mitochondrial potential and release of cytochrome c into the cytosol (5). Cytosolic Apaf-1 binds cytochrome c, and recruits and binds
pro-caspase-9; this assembly is known as the apoptosome (6). The apoptosome activates caspase-9 (7). Activated caspase-9 subsequently activates caspase-3/ −7 (8), leading to cell death.
(For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)

4.2.1. Bak/Bax and mitochondrial depolarization and outer membranes is termed the intermembrane space (IMS)
The mitochondrion is essential for regulating cell survival. Its ul- (Fig. 3A) [20]. The OM allows passage of specific cytosolic molecules
trastructure consists of a central matrix surrounded by an inner mem- through the voltage-dependent anion channel (VDAC). The IM contains
brane (IM) and an outer membrane (OM). The space between the inner a complex of proteins, called the electron transport chain (ETC), that

Fig. 3. Mitochondria-mediated apoptosis. A. The mitochondrial ultrastructure consists of the matrix, the inner membrane (IM), outer membrane (OM) and intermembrane space (IMS). B.
The mitochondrial permeability transition pore (MPTP) consists of the Voltage Dependent Anion Channel (VDAC), Adenine Nucleotide Translocator (ANT) and Cyclophilin D (CypD)
components. Under apoptotic conditions, the pore complex transports water and large molecules from the cytosol into the matrix. Consequently, the matrix swells and ruptures the IM and
OM. This results in the release of cytochrome c located in the intermembrane space (IMS). The MPTP can also lead to the dissipation of the H+ gradient that leads to mitochondrial
depolarization. C. Bak/Bax localized to the mitochondrial outer membrane (OM) can homo-oligomerize thus facilitating cytochrome c release.

3
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

donate/accept electrons to build a proton gradient between the matrix apoptotic signals such as DNA damage or ROS, activate MKK4/7 kinase,
and IMS [20]. This proton gradient maintains the mitochondrial elec- which then activates JNK. Activated JNK upregulates pro-apoptotic
trical potential characteristic of cells in their resting state. proteins such as Bad thus promoting mitochondrial depolarization and
Intrinsic apoptosis is associated with a marked increase in mi- apoptosis [37]. Similarly, p38 MAPK is activated by the MKK3/6 ki-
tochondrial membrane permeability, due to the formation of a mi- nase, whose purported downstream effects include cell cycle arrest and
tochondrial permeability transition pore (MPTP) complex (Fig. 3B). The apoptosis [36]. Finally, ERKs are activated upstream by phosphoryla-
MPTP permeabilizes the IM to the larger solutes of the IMS, which tion by MEK1/2. ERK activation can directly stimulate the mitochon-
eventually leads to the swelling of the matrix and loss of mitochondrial drial pathway of apoptosis, including cytochrome c release and caspase
potential (depolarization) [20,21]. This increased permeability also activation [38].
releases pro-apoptotic factors, namely cytochrome c, into the cytosol
[20,21]. Cytochrome c then binds to apoptotic protease-activating 4.2.5. Phosphoinositide 3-kinase (PI3K) signaling
factor (Apaf-1) to form a complex called the ‘apoptosome’ [20]. The PI3K phosphorylates the 3′ position of the inositol ring of phos-
apoptosome recruits and autoactivates caspase-9, which subsequently phoinositides, which activates phosphoinositide-dependent kinase 1
activates caspase-3 to execute cell death. (PDK1) kinase and protein kinase B (PKB) [39]. In nucleated cells, ac-
The MPTP complex is regulated by pro-apoptotic proteins, notably, tivated PDK1 phosphorylates and activates PKB, which promotes cell
activated Bax and Bak [22]. Activated Bax and Bak may homo-oligo- survival by phosphorylating and inactivating the pro-apoptotic protein
merize into pore complexes on the OM facilitating the release of mi- Bad [39].
tochondrial cytochrome c [21,23] (Fig. 3C). Consequently, intrinsic
apoptosis is also marked by MPTP formation, mitochondrial translo- 4.3. Caspase-independent apoptosis
cation of Bax, mitochondrial depolarization and cytochrome c release
[15]. A second group of inhibitory proteins, termed Bcl-2 proteins, bind While caspase activation is a well-documented endpoint of pro-
Bax and Bak to prevent their translocation to the mitochondria [14,24], apoptotic signaling, it should be noted that programmed platelet death
thus exerting an anti-apoptotic effect (Fig. 2). can also occur in a caspase-independent manner. Wolf and coworkers
noted that calcium ionophore treatment of platelets induces the cell
4.2.2. Calcium (Ca2+) signaling shrinkage, plasma membrane microvesiculation and PS externalization
Mitochondrial depolarization is also triggered by excessive accu- that are characteristic of apoptosis [40]. However, the authors also
mulation of intracellular Ca2+ that is transferred to the mitochondria noted that this Ca2+-driven apoptosis was mediated by the protease
[25,26]. Under apoptotic conditions, Ca2+ is released by the en- calpain and was not contingent on caspase activation [40]. In an in vitro
doplasmic reticulum (ER). Importantly, platelets contain an ER-like study of co-cultured platelets and macrophages, Brown et al docu-
dense tubular system (DTS) that serves as an intracellular Ca2+ store mented that aging platelets expressed increased Bak/Bax and PS ex-
[27]. Uptake of excess intracellular Ca2+ (Ca2+
i ) by the mitochondria posure but that the clearance of platelets by macrophages did not re-
leads to mitochondrial swelling and depolarization. In addition to an- quire caspase-3 activity [41]. Collectively, these findings indicate that
tagonizing the pro-apoptotic Bak and Bax proteins, the pro-survival Bcl- Ca2+ signaling fluxes can induce platelet apoptosis and clearance in-
2 proteins also prevent apoptosis by reducing the amount of releasable dependently of caspase activity.
Ca2+ from the ER and mitochondria [28]. A decreased Bcl-2/Bax (pro- Key points: apoptotic signaling in platelets (Fig. 2).
survival/pro-apoptotic) protein ratio was noted in platelets treated with
calcium ionophore [29], consistent with notion that Ca2+ influxes • During platelet apoptosis, the pro-apoptotic proteins Bak and Bax
trigger apoptosis. bind and permeabilize the mitochondrial membrane. The resultant
leaching of cytochrome c forms an Apaf-1/caspase-9 protein com-
4.2.3. Reactive oxygen species (ROS) plex (apoptosome) that activates caspase-3, causing cell death.
ROS are a family of oxygen-containing chemically reactive mole- • The pro-apoptotic activity of Bak and Bax are constrained by a group
cules. This family includes short-lived free radicals such as hydroxyl of proteins, termed “pro-survival” Bcl-2 proteins (Bcl-2, Bcl-xL).
(•OH), alkoxyl (RO•) or peroxyl (ROO•), and medium lifetime radicals • Another group of proteins known as BH3-only proteins (e.g. Bad,
such as superoxide (O2•) or nitroxyl radical (NO•). Other ROS include Bim, Bik, Puma), displace Bcl-2 proteins from Bak/Bax, thereby
hydrogen peroxide (H2O2), organic hydroperoxides (ROOH) and hy- promoting apoptosis.
pochlorous acid (HOCl) [30]. The accumulation of ROS in cells can • Apoptotic platelets typically exhibit mitochondrial depolarization,
promote excessive apoptosis. For example, ROS can target the mi- increased intracellular Ca2+ ([Ca2+]i) and surface PS exposure.
tochondria by oxidizing lipids on the mitochrondrial IM thereby en-
hancing cytochrome c release [31] and mitochondrial depolarization 5. Drugs with documented pro-apoptotic effects on platelets
[32]. Platelet apoptosis can be triggered both by platelet-derived or
vasculature-derived ROS [33]. Treating platelets with H2O2 evokes the More than 200 drugs are reported to cause thrombocytopenia
major hallmarks of apoptosis [27,34]. Conversely, inhibition of H2O2 [5,42]. The concept of platelet apoptosis is relatively new and was only
by catalase precludes PS exposure and caspase activation [27]. H2O2 recently evaluated in the context of drug-induced thrombocytopenia
treatment reportedly promotes the mitochondrial translocation of the [43]. Table 2 lists the drugs presently known to induce apoptosis in
pro-apoptotic proteins Bid and Bax [34]. ROS can also activate other platelets. Below we provide a detailed review of the drugs' putative pro-
signaling pathways, such as the mitogen activated protein kinase apoptotic mechanisms, based on currently available experimental evi-
(MAPK) pathways, which also have documented pro-apoptotic effects dence.
[35].
5.1. Acetylsalicylic acid (aspirin)
4.2.4. MAP kinase (MAPK) signaling
Mitogen-activated protein kinases (MAPK) signaling controls cell Aspirin is a common analgesic and anti-inflammatory drug used for
proliferation and survival [36]. MAPK signaling entails the sequential thrombosis prevention by 24–35% of adults over the age of 50 [44].
phosphorylation and activation of downstream target proteins. There The side effect of aspirin-induced thrombocytopenia was documented
are 3 MAPK modules associated with apoptosis: the c-Jun NH2 terminal as early as 1974 [45]. Recently published data by Zhao and colleagues
kinase (JNK) pathway; the p38 MAPK pathway; and the extracellular suggest that this may occur via apoptosis [46]. Using fluorescent dyes
signal regulated kinase (ERK) pathway. In the JNK pathway, pro- to measure mitochondrial potential and PS exposure in washed human

4
Table 2
Thrombocytopenia-inducing drugs with documented pro-apoptotic effects on platelets.

Drug class Drug name Clinical Indication Pro-apoptotic mechanism(s) of action Refs
E. De Silva, H. Kim

NSAID Aspirin Analgesia, anti-inflammatory, thrombosis prevention • Induction of mitochondrial depolarization, ROS generation and caspase-3 activation [46,47]
• Conformational modification of Bax that promotes Bax insertion into the outer mitochondrial
membrane
2+ 2+
Antibiotic Balhimycin Infection Increased intracellular Ca
• caspase-3 levels ([Ca ] ), induction of mitochondrial depolarization and
i [51]
activation
of pro-apoptotic ceramides
2+
Antibiotic Vancomycin Infection
• Formation
Increased [Ca ] , induction of mitochondrial depolarization, ceramide formation, increased
i [50]
• caspase-3 activity
2+ 2+
Antipsychotic Trifluoperazine Schizophrenia • Inhibition of calmodulin, thus inducing apoptosis by increasing [Ca ] and mitochondrial Ca
i [25]
overload
• Induction of mitochondrial depolarization, caspase-3 activation
Anti-neoplastic Carmustine Brain tumors • Induction of mitochondrial depolarization and caspase-3 activation [62]
• Upregulation of pro-apoptotic Bax; downregulation of anti-apoptotic Bcl-2
• c-Jun NH2-terminal kinase (JNK)-mediated mitochondrial depolarization
Anti-neoplastic ABT-737 (in development) • Main target Bcl-x < SUB > L < /SUB > [64,98–100]
• Induction of mitochondrial depolarization and release of cytochrome c, caspase-9, -8, -3 activation
Anti-neoplastic Cisplatin Various forms of cancer • Increased expression of pro-apoptotic Bax/Bak, decreased expression of anti-apoptotic Bcl-2/Bcl- [57,101]
X < SUB > L < /SUB >
• Increased binding of active Bax to mitochondrial membrane
of mitochondrial depolarization, caspase-3 activation, ROS production, increased
2+
• Induction
[Ca ] i
by MEK/ERK signaling but not p38 MAPK or JNK signaling
2+
Anti-neoplastic Tamoxifen Breast cancer
• Mediated of mitochondrial depolarization, caspase-3 activation, PS exposure, increased [Ca ]i [25]
Anti-neoplastic Navitoclax (ABT-263) (in development)
• Induction
with high affinity to Bcl2, Bcl-xL, Bcl-w, thus precluding their anti-apoptotic functions. [64,100]
2+
• Bind intracellular Ca by depletion of intracellular calcium stores (ER)

5
2+
Anti-neoplastic, immunosuppressant Methotrexate Hematologic cancers; rheumatoid arthritis
• Increased
generation, Ca release from ER, peroxidation of cardiolipin [59]
• ROS of mitochondrial depolarization, cytochrome c release, active caspase-9/ −3
• Induction expression of Bad, Bax, tBid; suppression of Bcl-2
• Increased
induces increased expression of Bad, Bax, suppression of anti-apoptotic Bcl-2 proteins,
• JNK
caspase-3 activation
2+
Anti-neoplastic NF-κB inhibitors Adenocarcinoma • ER stress, Ca release from ER, eIF2-α phosphorylation [67]
• MPTP formation, mitochondrial depolarization, cytochrome c release, caspase-3 activation, PS
exposure
• Decreased Bcl-2 levels, increased Bax levels
Anti-neoplastic Lovastatin Malignant glioma • Mitochondrial depolarization, upregulation of Bak, downregulation of Bcl-xL, caspase-3/-9 [71]
activation
• Integrin IIb 3
α β -mediated caspase-8 activation
Anti-neoplastic Doxorubicin Various forms of cancer • Mitochondrial translocation of Bax and mitochondrial depolarization, cytochrome c release, [76]
caspase-3 activation, PS exposure
• Augmented levels of mitochondrial ROS and cardiolipin peroxidation
Anti-neoplastic Bexarotene Colon, breast and lung cancer • Augmented thrombin and/or CRP stimulated intracellular calcium concentration [73]
• Caspase-3 IIb 3
activity, PS exposure and α β integrin activity
Anti-neoplastic Arsenic trioxide (ATO) Acute promyelocytic leukemia • Mitochondrial depolarization, upregulation of Bax, downregulation of Bcl-X < SUB > L < / [63]
SUB > , caspase-3 activation, PS exposure
• Induces apoptosis in a JNK-dependent manner
Food/consumable Ethanol Alcoholic beverages • Induction of mitochondrial depolarization and caspase-3 activation [83]
• Upregulation of Bax; downregulation of Bcl-2
of JNK
2+
Hormonal supplement Melatonin Purported benefits in regulating immunological,
• Activation ROS and H O levels, increase intracellular Ca
2 2 by depletion of intracellular stores [85]
cellular and endocrine responses
• Increased of mitochondrial depolarization, cytochrome c release, caspase-9 and -3 activity
2+
Natural derivative (black seed oil) Thymoquinone Purported benefits for multiple conditions, including
• Induction caspase-3 activity and [Ca ] < SUB > I < /SUB > [88]
cancer
• Increased of mitochondrial depolarization; increased pro-apoptotic ceramide formation
2+
• Induction i
• PI3K-mediated caspase-3 activation, increase in [Ca ] and mitochondrial depolarization (continued on next page)
Chemico-Biological Interactions 284 (2018) 1–11
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

platelets, the authors determined that aspirin induced platelet apoptosis


in a dose-dependent manner. Increased caspase-3 activation was also
[84,102]
noted in aspirin-treated platelets relative to controls. A separate study

[91]

[17]
Refs

documented that aspirin-treated platelets exhibit increased ROS gen-


caspase-3 and caspase-8 activity (not via classical death receptors), cleavage of Bid into eration and higher levels of the active form of the pro-apoptotic protein

of mitochondrial depolarization via both intrinsic and extrinsic pathways, caspase-9 and
Bax [47]. While aspirin's anti-thrombotic activity is attributed to the
caspase-9/-3 activity, induction of mitochondrial depolarization and cytochrome c

inhibition of cyclooxygenase (COX) synthesis [48], recent data raise the


possibility that aspirin's antithrombotic effects may be achieved by re-
• Bax translocation from cytosol to mitochondrial membrane, cytochrome c release ducing platelet numbers via apoptosis.

5.2. Antibiotics: vancomycin and balhimycin


ROS and H2O2 levels, increased [Ca ] < SUB > I < /SUB >

Antibiotics are another class of drugs associated with thrombocy-


topenia [49] although this was previously attributed to an antibody
-3 activation via caspase-8 activation, cleavage of Bid into tBid,

(hapten)-dependent mechanism [6] (Table 1). Two recent studies


evaluated the pro-apoptotic effects of antibiotics on human platelets
[50,51]. In these studies, treatment of platelets with vancomycin [50]
and balhimycin [51] significantly induced mitochondrial depolariza-
2+

tion, caspase-3 activation and PS exposure. Moreover, both antibiotics


stimulated the production of ceramides, which are lipids with docu-
of mitochondrial depolarization

mented pro-apoptotic functions [52]. In addition, increases in in-


tracellular [Ca2+] ([Ca2+]i) were observed in vancomycin- and bal-
Pro-apoptotic mechanism(s) of action

himycin-treated platelets loaded with the calcium indicator dye Fluo 3/


AM [50,51]. Interestingly, antibiotic-induced PS exposure, the endpoint
marker for apoptosis, was abrogated following calcium chelation with
tBid (BH3-only protein)

EDTA. However, PS exposure was not affected by caspase-3 inhibition


with zVAD-FMK, suggesting that [Ca2+]i influx, and not caspase-3 ac-
tivation, is the critical signaling event during antibiotic-induced platelet
apoptosis. These data suggest that vancomycin induces apoptosis in
• Induction
• Induction
• Increased
• Increased
• Increased

platelets in a caspase-independent manner (see section 4.3 above).


release

Further research is required to determine whether other antibiotics


induce thrombocytopenia through a comparable mechanism.

5.3. Anti-neoplastic agents

More than 100 chemotherapeutic drugs are currently employed as


anti-cancer agents [53]. Thrombocytopenia is a common side effect to
chemotherapy, due to the drugs' suppression of bone marrow and
thrombopoiesis [54]. However, emerging evidence published over the
Purported anti-oxidant properties

Purported anti-oxidant properties


Chronic inflammatory diseases

past 5 years suggests that chemotherapeutic agents may target circu-


lating platelets and induce their death by apoptosis.

5.3.1. Tamoxifen
Clinical Indication

Tamoxifen is a well-known chemotherapeutic used in the treatment


of breast cancer [55]. Tamoxifen-treated human platelets reportedly
exhibit increased mitochondrial depolarization and PS exposure; the
drug's antagonism of calmodulin (CaM) was proposed as the mechanism
of action [25]. CaM is a Ca2+-binding protein that modulates Ca2+-
dependent signaling processes [56] and also regulates cytosolic calcium
concentration [25]. These data suggest that tamoxifen induces in-
Andrographolide

tracellular Ca2+ fluxes by antagonizing CaM, thus promoting platelet


apoptosis.
Resveratrol
Drug name

Sesamol

5.3.2. Cisplatin
A more complete mechanistic description of drug-induced apoptosis
was provided for the anti-cancer agent cisplatin [57]. Consistent with
Natural derivative (plant-derived)

Natural derivative (plant-derived)

the findings reported in other studies of washed human platelets, cis-


Natural derivative (sesame seeds)

platin treatment induced mitochondrial depolarization, caspase-3 acti-


vation, Ca2+
i influx and PS exposure [57]. Moreover, as was observed in
vancomycin-induced apoptosis [50], cisplatin-driven platelet apoptosis
was shown to be Ca2+-dependent. However, the authors reported four
Table 2 (continued)

(4) additional features of cisplatin-induced platelet apoptosis. Firstly,


immunoblots of cisplatin-treated platelets showed increased expression
Drug class

of the pro-apoptotic Bax and Bak proteins relative to untreated controls.


Conversely, levels of the anti-apoptotic proteins Bcl-2 and Bcl-XL are
attenuated following cisplatin exposure. Secondly, immunoblot analysis

6
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

of mitochondrial fractions revealed increased mitochondria-associated drugs promoted increased [Ca2+]i, and eIF2α phosphorylation [67].
Bax in cisplatin-treated platelets, indicating that cisplatin drives the Platelets treated with BAY11-7082 and MLN4924 also exhibited, in a
translocation of Bax to the mitochondria. Thirdly, the authors identified dose-dependent manner, decreased Bcl-2 expression, increased Bax
the MEK/ERK signaling pathway as the target for cisplatin-induced expression, MPTP formation, cytochrome c release and caspase-3 acti-
apoptosis, since mitochondrial depolarization, caspase-3 activation and vation. Importantly, the mitochondrial depolarization and PS exposure
PS exposure were abrogated in platelets pre-treated with the MEK in- induced by the NF-κB inhibitors were rescued by incubating the pla-
hibitor PD98059 [57]. Fourthly, cisplatin reportedly promotes ROS telets with an ER stress inhibitor (Salubrinal), further supporting the
generation by platelets, as quantified by a fluorescent ROS-labeling dye; notion that NF-κB inhibition drives platelet apoptosis via ER stress [67].
interestingly, quenching of ROS by a reducing agent (DTT) also nulli-
fied cisplatin-induced apoptosis [57]. Consequently, this study provides 5.3.6. Lovastatin
a thorough description of a drug-induced apoptotic pathway that is Lovastatin is a member of the statin family of drugs used to lower
simultaneously dependent on ROS generation, as well as Ca2+ and blood cholesterol, with additional applications as an anti-cancer ther-
MEK/ERK signaling. apeutic [69,70]. Recently published data indicate that washed human
platelets treated with lovastatin displayed dose-dependent increases in
5.3.3. Methotrexate, carmustine and arsenic trioxide (ATO) mitochondrial depolarization, upregulation of Bak, downregulation of
Methotrexate is an anti-cancer drug that is also used to treat rheu- Bcl-xL and activated caspase-3/-9, relative to controls [71]. A notable
matoid arthritis [58] and whose role in platelet apoptosis was recently finding from of this study was the abrogation of mitochondrial depo-
documented [59]. Methotrexate treatment induced endogenous ROS larization in the presence of an αIIbβ3 integrin-blocking peptide, sug-
production in platelets, as previously reported with cisplatin [57]. Mi- gesting that lovastatin targets the αIIbβ3 integrin complex (also termed
tochondrial depolarization, increased [Ca2+]i, caspase-3 activation and glycoprotein IIbIIIa) to induce platelet apoptosis since [71]. These data
PS exposure were all observed in methotrexate-treated platelets [59]. In provide additional evidence in support of receptor-mediated platelet
this study, methotrexate induced JNK phosphorylation; this effect was apoptosis that is independent of the classical Fas death receptors.
nullified by the JNK inhibitor dicumarol as well as the ROS antagonist
Mito-TEMPO. Further, methotrexate treatment upregulated the ex- 5.3.7. Bexarotene
pression of the pro-apoptotic Bad and Bax proteins while down- Bexarotene is a stimulator of retinoid X receptors that is used as a
regulating expression of the anti-apoptotic Bcl-2. These findings were chemotherapeutic agent against several different types of cancer [72].
reversed following JNK inhibition, thus indicating that methotrexate The pro-apoptotic effects of bexarotene were recently documented in
induces platelet apoptosis by specifically targeting the JNK pathway in mouse platelets stimulated concurrently with thrombin or collagen-re-
a ROS-dependent manner [59]. Similar pro-apoptotic mechanisms were lated peptide (CRP) [73]. Cao et al found that thrombin and CRP in-
reported for carmustine, an alkylating chemotherapeutic used to treat duced caspase-3 activity, [Ca2+]i and PS exposure, and that these ef-
several different cancers, including brain tumors and lymphomas [60] fects were significantly enhanced in the presence of bexarotene [73].
as well as for arsenic trioxide (ATO), a highly potent anti-neoplastic Interestingly, these data also suggest that bexarotene, like lovastatin,
drug used to treat acute promyelocytic leukemia [61]. As observed with targets the αIIbβ3 integrin complex (GPIIbIIIa), since CRP-induced
other drugs, carmustine and ATO induce mitochondrial depolarization, αIIbβ3 activation was slightly increased in the presence of bexarotene
upregulation of Bax, downregulation of Bcl-2 and caspase-3 activation [73].
in human platelets [62,63]. Like methotrexate, carmustine- and ATO-
induced mitochondrial depolarization are reportedly nullified in the 5.3.8. Doxorubicin
presence of dicumarol, and thus appear to be JNK-dependent [62,63]. Doxorubicin (DOX) is another chemotherapeutic drug used for the
treatment of multiple forms of cancer [74]. As is observed with many
5.3.4. Bcl-2-inhibiting drugs anti-neoplastic agents, DOX is associated with thrombocytopenia [75].
Another class of anti-cancer drugs target the anti-apoptotic Bcl-2 In a report by Wang and colleagues, DOX-treated platelets exhibited
proteins, which bind and inhibit the pro-apoptotic proteins Bak and Bax increased mitochondrial translocation of Bax and mitochondrial depo-
[24]. ABT-737, and its oral analogue, ABT-263 (Navitoclax), bind to larization, which was also associated with cytochrome c release, cas-
Bcl-2, thus nullifying its anti-apoptotic effect. Vogler et al observed that pase-3 activation, PS exposure and increased ROS generation [76].
ABT-737 and ABT-263 induced mitochondrial depolarization, increased Importantly, DOX-induced apoptotic markers in platelets were in-
[Ca2+]i caspase-3 activation and PS exposure in platelets [64]. Another hibited by Mito-TEMPO, a mitochondrial reactive oxygen species (ROS)
group reported that ABT-737 activates the p38 MAP kinase pathway in antagonist [76]. These data imply that mitochondrial ROS play a cen-
platelets, and that drug-induced mitochondrial depolarization, caspase- tral role in DOX-induced platelet apoptosis.
3 activation and PS exposure were all attenuated by SB202190, a p38
MAPK inhibitor [65]. In addition, it was noted that ABT-737-induced 5.4. Food-derived, natural and herbal substances
platelet apoptosis is caspase-3-dependent, in contrast with the caspase-
3-independent mechanism observed with vancomycin-induced platelet Alternative medicines, including herbal medicines, are commonly
apoptosis [50]. Collectively, these data underscore the heterogeneous, used to manage symptoms associated with chronic pain [77], ulcerative
multi-pronged (and possibly drug-specific) nature of pro-apoptotic colitis [78] and even menopause [79]. As is the case with medications
signaling pathways (Fig. 4). prescribed by health care providers, these natural remedies may confer
significant side effects, such as thrombocytopenia [80]. Published data
5.3.5. Nuclear factor kappa B (NF-κB) inhibitors from the past 5 years demonstrate the ability of several naturally-de-
The NF-κB family of proteins regulate gene transcription and pro- rived products to induce apoptosis in platelets.
mote cell survival; consequently, the NF-κB signaling pathway is a
target for cancer therapy [66]. Recent evidence suggests that NF-κB 5.4.1. Ethanol
inhibition via the drugs BAY11-7082 and MLN4924 induces platelet Chronic alcohol use has long been associated with an increased
apoptosis by promoting endoplasmic reticulum (ER) stress [67]. susceptibility for bleeding and thrombocytopenia [81,82]. Newly pub-
ER stress is characterized by the accumulation of unfolded proteins lished work identifies ethanol as a pro-apoptotic stimulus for platelets
in the ER whose downstream effects include apoptosis [68] and phos- [83]. Washed platelets treated with ethanol exhibit increases in mi-
phorylation of eukaryotic initiation factor 2 alpha (eIF2α). Paul et al tochondrial depolarization, caspase-3 activation, as well as up- and
evaluated platelets treated with NF-κB inhibitors and found that these down-regulation of Bax and Bcl-2, respectively [83]. The authors also

7
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

Fig. 4. An overview of known platelet apoptosis-inducing drugs and pertinent signaling pathways. The extrinsic pathway includes caspase-8 activation, which can be induced by ABT-
737, andrographolide, lovastatin and resveratrol. The intrinsic pathway is defined by several key features, which can be induced by the following drugs: up-regulation of Bax expression
induced by aspirin, doxorubicin, Navitoclax, ethanol, ABT-737 and resveratrol; ROS production induced by aspirin, doxorubicin, sesamol, melatonin and methotrexate. Increased
intracellular calcium concentration ([Ca2+]i) is induced by methotrexate, tamoxifen, balhimycin, vancomycin, Navitoclax, melatonin, sesamol and bexarotene. ER-stress mediated
apoptosis is induced by NF-κB inhibitors. Cisplatin targets the intrinsic pathway via ERK signaling; JNK signaling is induced by carmustine, methotrexate and arsenic trioxide; PI3K
signaling is induced by thymoquinone. Legend: Blue boxes highlight the key markers of the extrinsic and intrinsic apoptosis pathways; green boxes represent the major signaling pathways
(e.g. MAPK, PI3K and ER-stress) that promote platelet apoptosis. (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this
article.)

report an ethanol-induced increase in JNK activation in platelets, as was 5.4.4. Andrographolide and resveratrol: evidence for extrinsic apoptosis in
previously reported for methotrexate [59] and carmustine [62]. platelets
Moreover, the administration of ethanol to mice reduced their circu- Andrographolide is a compound derived from the plant
lating platelet counts in vivo and also increased bleeding times [83]. Andrographis paniculata that is employed as a herbal medicine for its
These data provide a partial explanation for the thrombocytopenia purported anti-inflammatory and anti-cancer properties [90]. Andro-
observed with chronic alcohol use. grapholide was reported to induce caspase-3 as well as caspase-8 acti-
vation in human platelets [91]. Similar data were obtained in a separate
study investigated the pro-apoptotic properties of resveratrol (3, 4′, 5-
5.4.2. Sesamol and melatonin trihydroxy-trans-stilbene), a polyphenol found in red wine [17]. In this
Sesamol is a phenolic compound contained in sesame seeds that study, resveratrol induced platelet apoptosis while exhibiting caspase-8
reportedly exhibits anti-neoplastic properties, but also promotes pla- activation, as was observed in andrographolide-treated platelets [17].
telet apoptosis [84]. Sesamol-treated platelets exhibited increased ROS The activation of caspase-8 by these drugs raises the novel and inter-
(H2O2) production, [Ca2+]i, mitochondrial depolarization, cytochrome esting concept that components of extrinsic apoptotic signaling may in
c release, caspase-3 and -9 activation and PS exposure [84]. The same fact exist in platelets.
group examined the pro-apoptotic effects of melatonin, an en- Key points: drug-induced platelet apoptosis.
dogenously-produced hormone available as an over-the-counter dietary
supplement [85]. Practically identical results were noted in melatonin- • Multiple drugs, including herbal substances, induce apoptosis in
treated platelets [85]. In addition, the authors noted that melatonin platelets.
treatment induced the phosphorylation of PI3-K, Syk and Src kinases • Most of the available evidence is limited to in vitro studies using
[85] although inhibition of these signaling pathways were not tested in washed platelets.
this study. • Apoptotic drug-treated platelets exhibit: (1) mitochondrial depo-
larization; (2) increased [Ca2+]i; (3) caspase-3 activation and (4) PS
exposure.
5.4.3. Thymoquinone • However, different drugs appear to trigger apoptosis via different
Thymoquinone is a chemical derived from the plant Nigela sativa mechanisms. These include: Bak activation, Bcl-2 inhibition, sti-
[86] that reportedly exhibits anti-cancer properties [87]. Thymoqui- mulation of different MAPK signaling modules (Fig. 4).
none treatment reportedly increases in [Ca2+]i, mitochondrial depo- • Drug-induced platelet apoptosis occurs mainly via receptor-in-
larization, caspase-3 activity and PS exposure [88,89]. These increases dependent (intrinsic) pathways, although some drugs induce cas-
were abrogated in the presence of the PI-3-kinase inhibitor wortmannin pase-8 activation, suggesting possible involvement of the extrinsic
[88], suggesting that thymoquinone-driven apoptosis is dependent on pathway.
PI-3-kinase signaling. This finding seemingly contradicts the anti-
apoptotic effect of PI-3-kinase signaling reported in nucleated cells
[39], suggesting that the downstream effects of certain signaling
pathways are cell-type specific.

8
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

6. Thrombocytopenia-inducing drugs: indirect evidence for pro- [2] E.M. Greenberg, E.S. Kaled, Thrombocytopenia, Crit. Care Nurs. Clin. 25 (2013)
apoptotic activity 427–434 v.
[3] B.H. Chong, P.Y. Choi, L. Khachigian, J. Perdomo, Drug-induced immune throm-
bocytopenia, Hematol. Oncol. Clin. N. Am. 27 (2013) 521–540.
Research from the past decade has revealed the importance of [4] G.P. Visentin, C.Y. Liu, Drug-induced thrombocytopenia, Hematol. Oncol. Clin. N.
apoptosis in controlling platelet lifespan [13,92]. We have also gleaned Am. 21 (2007) 685–696 (vi).
[5] R.H. Aster, B.R. Curtis, J.G. McFarland, D.W. Bougie, Drug-induced immune
considerable insight into apoptosis as a previously unidentified me- thrombocytopenia: pathogenesis, diagnosis, and management, J. Thromb.
chanism for drug-induced thrombocytopenia. This encourages spec- Haemostasis 7 (2009) 911–918.
ulation that other drugs known to cause platelet destruction, may in [6] B. Kenney, G. Stack, Drug-induced thrombocytopenia, Arch. Pathol. Lab Med. 133
(2009) 309–314.
fact do so by promoting platelet apoptosis. For example, interleukin-2 [7] P. Staibano, D.M. Arnold, D.M. Bowdish, I. Nazy, The unique immunological
(IL-2) therapy is associated with thrombocytopenia [93] and reportedly features of heparin-induced thrombocytopenia, Br. J. Haematol. 177 (2017)
induces apoptosis in thymocytes [94] but this has not been evaluated in 198–207.
[8] I. Michelmann, D. Bockmann, W. Nurnberger, S. Eckhof-Donovan, S. Burdach,
platelets. The platelet-destructive mechanisms of multiple drugs remain
U. Gobel, Thrombocytopenia and complement activation under recombinant TNF
unknown [9]. It would therefore be of considerable interest to evaluate alpha/IFN gamma therapy in man, Ann. Hematol. 74 (1997) 179–184.
the effects of these drugs in the context of platelet apoptosis. [9] A.D. Michelson, Platelets, third ed., Academic Press, London ; Waltham, MA, 2013.
[10] S. Elmore, Apoptosis: a review of programmed cell death, Toxicol. Pathol. 35
(2007) 495–516.
7. Perspectives [11] V. Nikoletopoulou, M. Markaki, K. Palikaras, N. Tavernarakis, Crosstalk between
apoptosis, necrosis and autophagy, Biochim. Biophys. Acta 1833 (2013)
7.1. Conclusion 3448–3459.
[12] A.V. Gyulkhandanyan, A. Mutlu, J. Freedman, V. Leytin, Markers of platelet
apoptosis: methodology and applications, J. Thromb. Thrombolysis 33 (2012)
Platelet apoptosis is a relatively new and understudied concept and 397–411.
there now exists convincing evidence that drugs cause thrombocyto- [13] B.T. Kile, The role of apoptosis in megakaryocytes and platelets, Br. J. Haematol.
165 (2014) 217–226.
penia via pro-apoptotic signaling. There is remarkable congruity across [14] M. Lebois, E.C. Josefsson, Regulation of platelet lifespan by apoptosis, Platelets 27
all studies with regards to the drugs' endpoint effects on Ca2+ signaling, (2016) 497–504.
mitochondrial depolarization and PS exposure in platelets. However, [15] V. Leytin, Apoptosis in the anucleate platelet, Blood Rev. 26 (2012) 51–63.
[16] K. Segawa, S. Nagata, An apoptotic 'eat me' signal: phosphatidylserine exposure,
the different drugs also target distinct signaling pathways (e.g. JNK vs. Trends Cell Biol. 25 (2015) 639–650.
ERK) (Fig. 4) and show other subtle differences in their modulation of [17] K.H. Lin, G. Hsiao, C.M. Shih, D.S. Chou, J.R. Sheu, Mechanisms of resveratrol-
apoptosis. For example, vancomycin-induced apoptosis is Ca2+-de- induced platelet apoptosis, Cardiovasc. Res. 83 (2009) 575–585.
[18] M.L. Kahn, M. Nakanishi-Matsui, M.J. Shapiro, H. Ishihara, S.R. Coughlin,
pendent but unaffected by caspase-3 blockade [50], whereas ABT-737
Protease-activated receptors 1 and 4 mediate activation of human platelets by
and thymoquinone induce apoptosis in a caspase-3-dependent manner thrombin, J. Clin. Invest. 103 (1999) 879–887.
[65,89]. It is tempting to hypothesize that drug-induced thrombocyto- [19] V. Leytin, D.J. Allen, S. Mykhaylov, E. Lyubimov, J. Freedman, Thrombin-trig-
penia could one day be mitigated by the concurrent administration of gered platelet apoptosis, J. Thromb. Haemostasis 4 (2006) 2656–2663.
[20] G. Kroemer, J.C. Reed, Mitochondrial control of cell death, Nat. Med. 6 (2000)
targeted anti-apoptotic agents, but this would obviously require a 513–519.
thorough and complete understanding of all drug-induced apoptotic [21] D.R. Green, J.C. Reed, Mitochondria and apoptosis, Science 281 (1998)
pathways. 1309–1312.
[22] S. Shimizu, M. Narita, Y. Tsujimoto, Bcl-2 family proteins regulate the release of
apoptogenic cytochrome c by the mitochondrial channel VDAC, Nature 399
7.2. Future directions (1999) 483–487.
[23] G. Dewson, R.M. Kluck, Mechanisms by which Bak and Bax permeabilise mi-
tochondria during apoptosis, J. Cell Sci. 122 (2009) 2801–2808.
A major limitation of the available evidence is that nearly all of the [24] M.A. Levy, D.F. Claxton, Therapeutic inhibition of BCL-2 and related family
studies were conducted in vitro using washed platelets. Future studies members, Expet Opin. Invest. Drugs 26 (2017) 293–301.
would need to directly evaluate the effect of drug administration on [25] Z. Wang, S. Li, Q. Shi, R. Yan, G. Liu, K. Dai, Calmodulin antagonists induce
platelet apoptosis, Thromb. Res. 125 (2010) 340–350.
circulating platelet counts in vivo. To thoroughly dissect the drug-in-
[26] M.R. Duchen, Mitochondria and calcium: from cell signalling to cell death, J.
duced apoptotic signaling pathways in vivo, future studies could employ Physiol. 529 (1) (2000) 57–68.
knockout mice where selected elements of putative signaling pathways [27] J.J. Lopez, G.M. Salido, E. Gomez-Arteta, J.A. Rosado, J.A. Pariente, Thrombin
induces apoptotic events through the generation of reactive oxygen species in
such as JNK or ERK are specifically deleted from platelets using Cre/lox
human platelets, J. Thromb. Haemostasis 5 (2007) 1283–1291.
technology [95]. These elegant approaches could also be combined [28] R. Rizzuto, P. Pinton, D. Ferrari, M. Chami, G. Szabadkai, P.J. Magalhaes, F. Di
with advanced imaging methods [96] to detect and analyze drug-in- Virgilio, T. Pozzan, Calcium and apoptosis: facts and hypotheses, Oncogene 22
duced apoptosis in animal models. Further studies in this area are thus (2003) 8619–8627.
[29] D.M. Vanags, S. Orrenius, M. Aguilar-Santelises, Alterations in Bcl-2/Bax protein
likely to yield valuable new knowledge regarding the molecular me- levels in platelets form part of an ionomycin-induced process that resembles
chanisms of drug function. apoptosis, Br. J. Haematol. 99 (1997) 824–831.
[30] H.U. Simon, A. Haj-Yehia, F. Levi-Schaffer, Role of reactive oxygen species (ROS)
in apoptosis induction, Apoptosis 5 (2000) 415–418.
Acknowledgements [31] P.S. Brookes, Y. Yoon, J.L. Robotham, M.W. Anders, S.S. Sheu, Calcium, ATP, and
ROS: a mitochondrial love-hate triangle, Am. J. Physiol. Cell Physiol. 287 (2004)
The authors thank all members of the Kim laboratory and the UBC C817–C833.
[32] S. Orrenius, V. Gogvadze, B. Zhivotovsky, Calcium and mitochondria in the reg-
Centre for Blood Research, for helpful discussions. This work was ulation of cell death, Biochem. Biophys. Res. Commun. 460 (2015) 72–81.
supported, in part, by a Canadian Institutes of Health Research (CIHR) [33] B. Wachowicz, J.Z. Rywaniak, P. Nowak, Apoptotic markers in human blood
(grant number MC2-127872) Clinician-Scientist Salary Award (to H·K.). platelets treated with peroxynitrite, Platelets 19 (2008) 624–635.
[34] J.J. Lopez, G.M. Salido, J.A. Pariente, J.A. Rosado, Thrombin induces activation
and translocation of Bid, Bax and Bak to the mitochondria in human platelets, J.
Transparency document Thromb. Haemostasis 6 (2008) 1780–1788.
[35] S.K. NaveenKumar, R.M. Thushara, M.S. Sundaram, M. Hemshekhar, M. Paul,
C. Thirunavukkarasu, Basappa, G. Nagaraju, S.C. Raghavan, K.S. Girish,
Transparency document related to this article can be found online at
K. Kemparaju, K.S. Rangappa, Unconjugated bilirubin exerts pro-apoptotic effect
http://dx.doi.org/10.1016/j.cbi.2018.01.015. on platelets via p38-MAPK activation, Sci. Rep. 5 (2015) 15045.
[36] T. Wada, J.M. Penninger, Mitogen-activated protein kinases in apoptosis regula-
References tion, Oncogene 23 (2004) 2838–2849.
[37] D.N. Dhanasekaran, E.P. Reddy, JNK signaling in apoptosis, Oncogene 27 (2008)
6245–6251.
[1] K. Eto, S. Kunishima, Linkage between the mechanisms of thrombocytopenia and [38] S. Cagnol, J.C. Chambard, ERK and cell death: mechanisms of ERK-induced cell
thrombopoiesis, Blood 127 (2016) 1234–1241. death–apoptosis, autophagy and senescence, FEBS J. 277 (2010) 2–21.

9
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

[39] V. Duronio, The life of a cell: apoptosis regulation by the PI3K/PKB pathway, [69] Y.K. Chae, M. Yousaf, M.K. Malecek, B. Carneiro, S. Chandra, J. Kaplan, A. Kalyan,
Biochem. J. 415 (2008) 333–344. A. Sassano, L.C. Platanias, F. Giles, Statins as anti-cancer therapy; Can we translate
[40] B.B. Wolf, J.C. Goldstein, H.R. Stennicke, H. Beere, G.P. Amarante-Mendes, preclinical and epidemiologic data into clinical benefit? Discov. Med. 20 (2015)
G.S. Salvesen, D.R. Green, Calpain functions in a caspase-independent manner to 413–427.
promote apoptosis-like events during platelet activation, Blood 94 (1999) [70] M.D. Johnson, A. Woodard, E.J. Okediji, S.A. Toms, G.S. Allen, Lovastatin is a
1683–1692. potent inhibitor of meningioma cell proliferation: evidence for inhibition of a
[41] S.B. Brown, M.C. Clarke, L. Magowan, H. Sanderson, J. Savill, Constitutive death mitogen associated protein kinase, J. Neuro Oncol. 56 (2002) 133–142.
of platelets leading to scavenger receptor-mediated phagocytosis. A caspase-in- [71] Q. Zhao, M. Li, M. Chen, L. Zhou, L. Zhao, R. Hu, R. Yan, K. Dai, Lovastatin induces
dependent cell clearance program, J. Biol. Chem. 275 (2000) 5987–5996. platelet apoptosis, Environ. Toxicol. Pharmacol. 42 (2016) 69–75.
[42] R.H. Aster, D.W. Bougie, Drug-induced immune thrombocytopenia, N. Engl. J. [72] K.T. Liby, M.B. Sporn, Rexinoids for prevention and treatment of cancer: oppor-
Med. 357 (2007) 580–587. tunities and challenges, Curr. Top. Med. Chem. (2016) [Epub ahead of print].
[43] R.M. Thushara, M. Hemshekhar, K. Kemparaju, K.S. Rangappa, S. Devaraja, [73] H. Cao, R. Bissinger, A.T. Umbach, M. Gawaz, F. Lang, Effect of bexarotene on
K.S. Girish, Therapeutic drug-induced platelet apoptosis: an overlooked issue in platelet activation and apoptosis, Cell. Physiol. Biochem. 42 (2017) 838–847.
pharmacotoxicology, Arch. Toxicol. 88 (2014) 185–198. [74] C. Chen, L. Lu, S. Yan, H. Yi, H. Yao, D. Wu, G. He, X. Tao, X. Deng, Autophagy and
[44] M. Stuntz, B. Bernstein, Recent trends in the prevalence of low-dose aspirin use for doxorubicin resistance in cancer, Anti Canc. Drugs 29 (2018) 1–9.
primary and secondary prevention of cardiovascular disease in the United States, [75] E.J. Kim, K.M. Lim, K.Y. Kim, O.N. Bae, J.Y. Noh, S.M. Chung, S. Shin, Y.P. Yun,
2012-2015, Prev. Med. Rep. 5 (2017) 183–186. J.H. Chung, Doxorubicin-induced platelet cytotoxicity: a new contributory factor
[45] S.K. Garg, C.R. Sarker, Aspirin-induced thrombocytopenia on an immune basis, for doxorubicin-mediated thrombocytopenia, J. Thromb. Haemostasis 7 (2009)
Am. J. Med. Sci. 267 (1974) 129–132. 1172–1183.
[46] L. Zhao, W. Zhang, M. Chen, J. Zhang, M. Zhang, K. Dai, Aspirin Induces platelet [76] Z. Wang, J. Wang, R. Xie, R. Liu, Y. Lu, Mitochondria-derived reactive oxygen
apoptosis, Platelets 24 (2013) 637–642. species play an important role in Doxorubicin-induced platelet apoptosis, Int. J.
[47] M.K. TranstransNayak, A. Dash, N. Singh, D. Dash, Aspirin delimits platelet life Mol. Sci. 16 (2015) 11087–11100.
span by proteasomal inhibition, PLoS One 9 (2014) e105049. [77] L. Chen, A. Michalsen, Management of chronic pain using complementary and
[48] M. Dovizio, A. Bruno, S. Tacconelli, P. Patrignani, Mode of action of aspirin as a integrative medicine, BMJ 357 (2017) j1284.
chemopreventive agent, Recent Results Canc. Res. 191 (2013) 39–65. [78] P. Wan, H. Chen, Y. Guo, A.P. Bai, Advances in treatment of ulcerative colitis with
[49] M.E. Johansen, J.U. Jensen, M.H. Bestle, L. Hein, A.O. Lauritsen, H. Tousi, herbs: from bench to bedside, World J. Gastroenterol. 20 (2014) 14099–14104.
K.M. Larsen, J. Loken, T. Mohr, K. Thormar, P.I. Johansson, A. Cozzi-Lepri, [79] O.H. Franco, R. Chowdhury, J. Troup, T. Voortman, S. Kunutsor, M. Kavousi,
J.D. Lundgren, The potential of antimicrobials to induce thrombocytopenia in C. Oliver-Williams, T. Muka, Use of plant-based therapies and menopausal
critically ill patients: data from a randomized controlled trial, PLoS One 8 (2013) symptoms: a systematic review and meta-analysis, J. Am. Med. Assoc. 315 (2016)
e81477. 2554–2563.
[50] S.T. Towhid, E.M. Schmidt, A. Tolios, P. Munzer, E. Schmid, O. Borst, M. Gawaz, [80] D.J. Royer, J.N. George, D.R. Terrell, Thrombocytopenia as an adverse effect of
E. Stegmann, F. Lang, Stimulation of platelet death by vancomycin, Cell. Physiol. complementary and alternative medicines, herbal remedies, nutritional supple-
Biochem. 31 (2013) 102–112. ments, foods, and beverages, Eur. J. Haematol. 84 (2010) 421–429.
[51] S.T. Towhid, A. Tolios, P. Munzer, E.M. Schmidt, O. Borst, M. Gawaz, E. Stegmann, [81] J.L. Heiman, Alcohol-induced thrombocytopenia: review of the literature and re-
F. Lang, Stimulation of platelet apoptosis by balhimycin, Biochem. Biophys. Res. port of case, J. Am. Dent. Assoc. 85 (1972) 1358–1361.
Commun. 435 (2013) 323–326. [82] J. Latvala, S. Parkkila, O. Niemela, Excess alcohol consumption is common in
[52] T.D. Mullen, L.M. Obeid, Ceramide and apoptosis: exploring the enigmatic con- patients with cytopenia: studies in blood and bone marrow cells, Alcohol Clin.
nections between sphingolipid metabolism and programmed cell death, Exp. Res. 28 (2004) 619–624.
Anticancer Agents Med Chem 12 (2012) 340–363. [83] L. Liu, M. Chen, L. Zhao, Q. Zhao, R. Hu, J. Zhu, R. Yan, K. Dai, Ethanol induces
[53] A.M. Burger, Highlights in experimental therapeutics, Canc. Lett. 245 (2007) platelet apoptosis, Alcohol Clin. Exp. Res. 41 (2017) 291–298.
11–21. [84] R.M. Thushara, M. Hemshekhar, K. Sunitha, M.S. Kumar, S. Naveen, K. Kemparaju,
[54] D. Daniel, J. Crawford, Myelotoxicity from chemotherapy, Semin. Oncol. 33 K.S. Girish, Sesamol induces apoptosis in human platelets via reactive oxygen
(2006) 74–85. species-mediated mitochondrial damage, Biochimie 95 (2013) 2060–2068.
[55] V.C. Jordan, Tamoxifen: a most unlikely pioneering medicine, Nat. Rev. Drug [85] K.S. Girish, M. Paul, R.M. Thushara, M. Hemshekhar, M. Shanmuga Sundaram,
Discov. 2 (2003) 205–213. K.S. Rangappa, K. Kemparaju, Melatonin elevates apoptosis in human platelets via
[56] M.W. Berchtold, A. Villalobo, The many faces of calmodulin in cell proliferation, ROS mediated mitochondrial damage, Biochem. Biophys. Res. Commun. 438
programmed cell death, autophagy, and cancer, Biochim. Biophys. Acta 1843 (2013) 198–204.
(2014) 398–435. [86] M.R. Khazdair, The protective effects of nigella sativa and its constituents on in-
[57] W. Zhang, L. Zhao, J. Liu, J. Du, Z. Wang, C. Ruan, K. Dai, Cisplatin induces duced neurotoxicity, J. Toxicol. 2015 (2015) 841823.
platelet apoptosis through the ERK signaling pathway, Thromb. Res. 130 (2012) [87] A.F. Majdalawieh, M.W. Fayyad, G.K. Nasrallah, Anti-cancer properties and me-
81–91. chanisms of action of thymoquinone, the major active ingredient of Nigella sativa,
[58] P.M. Brown, A.G. Pratt, J.D. Isaacs, Mechanism of action of methotrexate in Crit. Rev. Food Sci. Nutr. (0) (2017).
rheumatoid arthritis, and the search for biomarkers, Nat. Rev. Rheumatol. 12 [88] S.T. Towhid, E.M. Schmidt, E. Schmid, P. Munzer, S.M. Qadri, O. Borst, F. Lang,
(2016) 731–742. Thymoquinone-induced platelet apoptosis, J. Cell. Biochem. 112 (2011)
[59] M. Paul, M. Hemshekhar, R.M. Thushara, M.S. Sundaram, S.K. NaveenKumar, 3112–3121.
S. Naveen, S. Devaraja, K. Somyajit, R. West, Basappa S.C. Nayaka, U.I. Zakai, [89] N. Rukoyatkina, E. Butt, H. Subramanian, V.O. Nikolaev, I. Mindukshev,
G. Nagaraju, K.S. Rangappa, K. Kemparaju, K.S. Girish, Methotrexate promotes U. Walter, S. Gambaryan, P.M. Benz, Protein kinase A activation by the anti-cancer
platelet apoptosis via JNK-mediated mitochondrial damage: alleviation by N- drugs ABT-737 and thymoquinone is caspase-3-dependent and correlates with
Acetylcysteine and N-Acetylcysteine amide, PLoS One 10 (2015) e0127558. platelet inhibition and apoptosis, Cell Death Dis. 8 (2017) e2898.
[60] M. Sabel, A. Giese, Safety profile of carmustine wafers in malignant glioma: a [90] J.C. Lim, T.K. Chan, D.S. Ng, S.R. Sagineedu, J. Stanslas, W.S. Wong,
review of controlled trials and a decade of clinical experience, Curr. Med. Res. Andrographolide and its analogues: versatile bioactive molecules for combating
Opin. 24 (2008) 3239–3257. inflammation and cancer, Clin. Exp. Pharmacol. Physiol. 39 (2012) 300–310.
[61] C.H. Ng, W.J. Chng, Recent advances in acute promyelocytic leukaemia, F1000Res [91] L.M. Lien, C.C. Su, W.H. Hsu, W.J. Lu, C.L. Chung, T.L. Yen, H.C. Chiu, J.R. Sheu,
6 (2017) 1273. K.H. Lin, Mechanisms of andrographolide-induced platelet apoptosis in human
[62] J. Zhang, M. Chen, Y. Zhang, L. Zhao, R. Yan, K. Dai, Carmustine induces platelet platelets: regulatory roles of the extrinsic apoptotic pathway, Phytother Res. 27
apoptosis, Platelets 26 (2015) 437–442. (2013) 1671–1677.
[63] Y. Wu, J. Dai, W. Zhang, R. Yan, Y. Zhang, C. Ruan, K. Dai, Arsenic trioxide in- [92] M. Lebois, M.R. Dowling, P. Gangatirkar, P.D. Hodgkin, B.T. Kile, W.S. Alexander,
duces apoptosis in human platelets via C-Jun NH2-terminal kinase activation, E.C. Josefsson, Regulation of platelet lifespan in the presence and absence of
PLoS One 9 (2014) e86445. thrombopoietin signaling, J. Thromb. Haemostasis 14 (2016) 1882–1887.
[64] M. Vogler, H.A. Hamali, X.M. Sun, E.T. Bampton, D. Dinsdale, R.T. Snowden, [93] E.R. Bregani, P. Lissoni, F. Rossini, S. Barni, G. Tancini, F. Brivio, A. Conti,
M.J. Dyer, A.H. Goodall, G.M. Cohen, BCL2/BCL-X(L) inhibition induces apop- G.J. Maestroni, Prevention of interleukin-2-induced thrombocytopenia during the
tosis, disrupts cellular calcium homeostasis, and prevents platelet activation, immunotherapy of cancer by a concomitant administration of the pineal hormone
Blood 117 (2011) 7145–7154. melatonin, Recenti Prog. Med. 86 (1995) 231–233.
[65] N. Rukoyatkina, I. Mindukshev, U. Walter, S. Gambaryan, Dual role of the p38 [94] G. Migliorati, I. Nicoletti, M.C. Pagliacci, L. D'Adamio, C. Riccardi, Interleukin-2
MAPK/cPLA2 pathway in the regulation of platelet apoptosis induced by ABT-737 induces apoptosis in mouse thymocytes, Cell. Immunol. 146 (1993) 52–61.
and strong platelet agonists, Cell Death Dis. 4 (2013) e931. [95] R. Tiedt, T. Schomber, H. Hao-Shen, R.C. Skoda, Pf4-Cre transgenic mice allow the
[66] S.A. Vlahopoulos, O. Cen, N. Hengen, J. Agan, M. Moschovi, E. Critselis, generation of lineage-restricted gene knockouts for studying megakaryocyte and
M. Adamaki, F. Bacopoulou, J.A. Copland, I. Boldogh, M. Karin, G.P. Chrousos, platelet function in vivo, Blood 109 (2007) 1503–1506.
Dynamic aberrant NF-kappaB spurs tumorigenesis: a new model encompassing the [96] F.G. Blankenberg, In vivo detection of apoptosis, J. Nucl. Med. 49 (2) (2008)
microenvironment, Cytokine Growth Factor Rev. 26 (2015) 389–403. 81S–95S.
[67] M. Paul, K. Kemparaju, K.S. Girish, Inhibition of constitutive NF-kappaB activity [97] T. Kam, M. Alexander, Drug-induced immune thrombocytopenia, J. Pharm. Pract.
induces platelet apoptosis via ER stress, Biochem. Biophys. Res. Commun. 493 27 (2014) 430–439.
(2017) 1471–1477. [98] A.V. Gyulkhandanyan, A. Mutlu, J. Freedman, V. Leytin, Mitochondrial perme-
[68] M. Kitamura, Endoplasmic reticulum stress and unfolded protein response in renal ability transition pore (MPTP)-dependent and -independent pathways of mi-
pathophysiology: janus faces, Am. J. Physiol. Ren. Physiol. 295 (2008) tochondrial membrane depolarization, cell shrinkage and microparticle formation
F323–F334. during platelet apoptosis, Br. J. Haematol. 169 (2015) 142–145.

10
E. De Silva, H. Kim Chemico-Biological Interactions 284 (2018) 1–11

[99] A. Mutlu, A.V. Gyulkhandanyan, J. Freedman, V. Leytin, Activation of caspases-9, Chemotherapy-induced thrombocytopenia derives from the selective death of
-3 and -8 in human platelets triggered by BH3-only mimetic ABT-737 and calcium megakaryocyte progenitors and can be rescued by stem cell factor, Canc. Res. 67
ionophore A23187: caspase-8 is activated via bypass of the death receptors, Br. J. (2007) 4767–4773.
Haematol. 159 (2012) 565–571. [102] R.M. Thushara, M. Hemshekhar, M. Paul, M. Shanmuga Sundaram, R.L. Shankar,
[100] Y. Harazono, K. Nakajima, A. Raz, Why anti-Bcl-2 clinical trials fail: a solution, K. Kemparaju, K.S. Girish, Crocin prevents sesamol-induced oxidative stress and
Canc. Metastasis Rev. 33 (2014) 285–294. apoptosis in human platelets, J. Thromb. Thrombolysis 38 (2014) 321–330.
[101] A. Zeuner, M. Signore, D. Martinetti, M. Bartucci, C. Peschle, R. De Maria,

11

You might also like