ArticlePDF Available

Novel inosine monophosphate dehydrogenase inhibitor VX-944 induces apoptosis in multiple myeloma cells primarily via caspase-independent AIF/Endo G pathway

Authors:

Abstract and Figures

Inosine monophosphate dehydrogenase (IMPDH) is a rate-limiting enzyme required for the de novo synthesis of guanine nucleotides from IMP. VX-944 (Vertex Pharmaceuticals, Cambridge, MA, USA) is a small-molecule, selective, noncompetitive inhibitor directed against human IMPDH. In this report, we show that VX-944 inhibits in vitro growth of human multiple myeloma (MM) cell lines via induction of apoptosis. Interleukin-6, insulin-like growth factor-1, or co-culture with bone marrow stromal cells (BMSCs) do not protect against VX-944-induced MM cell growth inhibition. VX-944 induced apoptosis in MM cell lines with only modest activation of caspases 3, 8, and 9. Furthermore, the pan-caspase inhibitor z-VAD-fmk did not inhibit VX-944-induced apoptosis and cell death. During VX-944-induced apoptosis, expressions of Bax and Bak were enhanced, and both apoptosis-inducing factor (AIF) and endonuclease G (Endo G) were released from the mitochondria to cytosol, suggesting that VX-944 triggers apoptosis in MM cells primarily via a caspase-independent, Bax/AIF/Endo G pathway. Importantly, VX-944 augments the cytotoxicity of doxorubicin and melphalan even in the presence of BMSCs. Taken together, our data demonstrate a primarily non-caspase-dependent apoptotic pathway triggered by VX-944, thereby providing a rationale to enhance MM cell cytotoxicity by combining this agent with conventional agents which trigger caspase activation.
Content may be subject to copyright.
Novel inosine monophosphate dehydrogenase inhibitor VX-944 induces
apoptosis in multiple myeloma cells primarily via caspase-independent
AIF/Endo G pathway
Kenji Ishitsuka
1
, Teru Hideshima
1
, Makoto Hamasaki
1
, Noopur Raje
1
, Shaji Kumar
1
,
Klaus Podar
1
, Steven Le Gouill
1
, Norihiko Shiraishi
1
, Hiroshi Yasui
1
, Aldo M Roccaro
1
,
Yu-Zu Tai
1
, Dharminder Chauhan
1
, Robert Fram
2
, Kazuo Tamura
3
, Jugnu Jain
2
and
Kenneth C Anderson*
,1
1
Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical
School, 44 Binney Street, Boston, MA 02115, USA;
2
Vertex Pharmaceuticals Inc., 130 Waverly Street, Cambridge, MA 02139, USA;
3
1st Department of Internal Medicine, Fukuoka University, 7-45-1 Nanakuma Jonan Fukuoka 814-0180, Japan
Inosine monophosphate dehydrogenase (IMPDH) is a
rate-limiting enzyme required for the de novo synthesis of
guanine nucleotides from IMP. VX-944 (Vertex Pharma-
ceuticals, Cambridge, MA, USA) is a small-molecule,
selective, noncompetitive inhibitor directed against human
IMPDH. In this report, we show that VX-944 inhibits
in vitro growth of human multiple myeloma (MM) cell
lines via induction of apoptosis. Interleukin-6, insulin-like
growth factor-1, or co-culture with bone marrow stromal
cells (BMSCs) do not protect against VX-944-induced
MM cell growth inhibition. VX-944 induced apoptosis
in MM cell lines with only modest activation of caspases
3, 8, and 9. Furthermore, the pan-caspase inhibitor
z-VAD-fmk did not inhibit VX-944-induced apoptosis
and cell death. During VX-944-induced apoptosis, expres-
sions of Bax and Bak were enhanced, and both apoptosis-
inducing factor (AIF) and endonuclease G (Endo G) were
released from the mitochondria to cytosol, suggesting that
VX-944 triggers apoptosis in MM cells primarily via a
caspase-independent, Bax/AIF/Endo G pathway. Impor-
tantly, VX-944 augments the cytotoxicity of doxorubicin
and melphalan even in the presence of BMSCs. Taken
together, our data demonstrate a primarily non-caspase-
dependent apoptotic pathway triggered by VX-944,
thereby providing a rationale to enhance MM cell
cytotoxicity by combining this agent with conventional
agents which trigger caspase activation.
Oncogene (2005) 24, 5888–5896. doi:10.1038/sj.onc.1208739;
published online 6 June 2005
Keywords: multiple myeloma; IMPDH inhibitor;
VX-944; apoptosis; AIF/Endo G pathway
Introduction
Multiple myeloma (MM) remains incurable despite
conventional therapies including high-dose chemother-
apy with stem cell support. Importantly, novel agents
like thalidomide, the immunomodulatory drug Revli-
mid, and the proteasome inhibitor bortezomib can
achieve responses in patients with relapsed and refrac-
tory MM, and are now under evaluation as treatment
for patients earlier in their disease course (Richardson
et al., 2002, 2003, 2004). Nonetheless, there remains an
urgent need for the development of new therapeutic
strategies for MM treatment.
Inosine monophosphate dehydrogenase (IMPDH) is
a rate-limiting enzyme required for the de novo synthesis
of guanine nucleotides from IMP (Zimmermann et al.,
1998). There are two IMPDH isoforms in mammalian
cells: type I is constitutively expressed in most cell types,
and catalytically similar type II is activated in prolifer-
ating cells (Nagai et al., 1991; Senda and Natsumeda,
1994). Gene expression of IMPDH type II is elevated in
patient MM cells compared with normal control plasma
cells (Takebe et al., 2004). Conversely, inhibition of
IMPDH induces depletion of guanine nucleotide pools,
followed by decreased DNA and RNA synthesis
(Laliberte et al., 1998). Recently, IMPDH has been
shown to bind nucleic acids in vitro and in vivo,
suggesting that it may have a direct role in replication
and transcription (McLean et al., 2004). IMPDH
inhibitors induce cell-cycle arrest and decrease T- and
B-cell responses effectively, both in vitro and in vivo.
Consequently, they have been evaluated primarily as
immunosuppressive rather than anticancer therapies
(Eugui and Almquist, 1990; Eugui et al., 1991; Turka
et al ., 1991; Jain et al., 2001). Specifically, the IMPDH
inhibitors benzamide riboside (BR) and mycophenolic
acid (MPA) were found to induce growth inhibition, but
not cytotoxicity, in the panel of 60 cancer cell lines
derived from hematological and solid tumors at the
National Cancer Institute (http://dtp.nci.nih.gov). More
recently, however, BR and MPA have been shown to
Received 28 December 2004; revised 22 March 2005; accepted 22 March
2005; published online 6 June 2005
*Correspondence: KC Anderson;
E-mail: kenneth_anderson@dfci.harvard.edu
Oncogene (2005) 24, 5888–5896
&
2005 Nature Publishing Group
All rights reserved 0950-9232/05 $30.00
www.nature.com/onc
induce apoptosis in selected cancer cells (Hunakova
et al., 2000; Szekeres et al., 2002; Yalowitz et al., 2002;
Messina et al., 2004). Although activation of caspases
and inhibition of PARP have been reported as possible
mediators of apoptosis triggered by MPA and BR
(Yalowitz and Jayaram, 2002; Gu et al., 2003), their
mechanisms of action are not fully characterized. MPA
and tiazofurin, another IMPDH inhibitor, have been
evaluated in cancer animal models (Sweeney et al., 1972;
Ahluwalia et al., 1984), as well as in clinical trials, for
the treatment of leukemia and CML, respectively
(Knudtzon and Nissen, 1972; Wright et al., 1996). Both
compounds showed some objective responses, but
gastrointestinal intolerance (MPA) or neurotoxicity
(tiazofurin) has limited further investigation. Hence,
evaluation of more selective and well-tolerated IMPDH
inhibitors is needed to determine the therapeutic
potential of this approach in the treatment of malig-
nancy.
VX-944 (Vertex Pharmaceuticals, Cambridge, MA,
USA) is a small-molecule, selective, noncompetitive
inhibitor of both human IMPDH isoforms (Ki
6–10 n
M). In this study, we evaluated the cytotoxicity
of VX-944 against MM cells in vitro. We demonstrate
that VX-944 inhibits growth of MM cell lines by
induction of apoptosis, primarily via a caspase-indepen-
dent pathway.
Results
VX-944 inhibits growth of MM cell lines
The effect of VX-944 on growth of MM cell lines was
determined using the MTT assay. VX-944 significantly
inhibited the growth of RPMI8226, MM.1S, and U266
cells in a dose-dependent fashion, with 50% inhibition
(IC
50
) values at 48 h of 450, 450, and 600 nM,
respectively. VX-944 also inhibited growth of drug-
resistant cell lines, including doxorubicin (Dox)-resistant
RPMI8226-Dox40, melphalan (Mel) resistant
RPMI8226-LR5, and Dex (dexamethasone) resistant
MM.1R cells, with IC
50
values similar to the parental
drug-sensitive cell lines (Figure 1a). The degree of
growth inhibition at 72 h was observed to be similar to
that at 48 h. The effects of VX-944 on DNA synthesis
were also determined by measuring [
3
H]thymidine
incorporation during the last 8 h of 48-h cultures. IC
50
values determined by this method were lower than those
determined in the MTT assay: 350 n
M for RPMI8226,
100 n
M for MM.1S and 75 nM for U266 cells (data not
shown). Growth inhibition induced by VX-944 is
significantly reversed by the addition of guanosine,
which is converted to guanine and thereby circumvents
the effect of inhibition of IMPDH on guanine nucleotide
levels (Zimmermann et al ., 1998) (Figure 1b). Impor-
tantly, VX-944 did not reduce the viability of peripheral
blood mononuclear cells (PBMNCs) derived from
healthy individuals at 48 h at concentrations up to
6400 n
M (Figure 1c). This is consistent with the
expression of type II IMPDH, which is lower in normal
lymphocytes as compared to cancer cells. Increased
expression of type II isoform has been closely linked
with an increase in total IMPDH activity in cancer cells
(Nagai et al., 1991).
VX-944 induces caspase-independent apoptosis
in MM cells
In order to further characterize the cytotoxic effects of
VX-944 on MM cell lines, apoptosis induced by VX-944
was analysed. ANNEXIN V/PI staining showed 13.4
and 16.8% of ANNEXIN V þ /PIcells, as well as 40.4
and 55.5% of ANNEXIN V þ /PI þ cells, after 72 h-
exposure to 800 n
M VX-944 in RPMI8226 and MM.1S
cells, respectively (Figure 2a). VX-944-induced apopto-
sis was further confirmed by TUNEL assay cultured for
96 h with 800 n
M of VX-944 (Figure 2b).
Figure 1 Effects of VX-944 on proliferation of MM cell lines and
viability of normal PBMNCs, assessed by MTT assay after 48 h
culture. VX-944 inhibited proliferation of MM cell lines in a dose-
dependent manner (a); conversely, addition of guanosine signifi-
cantly reversed the inhibitory effects of VX-944 in RPMI8226 cells
(b). VX-944 did not reduce the viability of PBMNCs derived from
three healthy subjects (c)
VX-944 induces apoptosis in MM cells
K Ishitsuka et al
5889
Oncogene
We next examined the apoptotic pathway induced by
VX-944. Cleavage of caspase-8, -9, and -3 triggered
by VX-944 in MM.1S and RPMI8226 cells was modest,
and there was no cleavage of DFF45 (Figure 3a).
Caspases 6 and 7, mediators of caspase 3-independent
apoptosis (Miyashita et al., 1998; Kagawa et al., 2001;
Pirnia et al., 2002), were not activated by VX-944 (data
not shown). To further confirm caspase-independent
Figure 2 Effects of VX-944 on induction of apoptosis. Induction of apoptosis induced by VX-944 (800 nM) was shown by ANNEXIN
V-PI staining at 48 and 72 h (a) and TUNEL assay at 96 h (b)
VX-944 induces apoptosis in MM cells
K Ishitsuka et al
5890
Oncogene
apoptosis, the effects of z-VAD-fmk on cytotoxicity
and apoptosis induced by VX-944 were next determined.
Z-VAD-fmk is a broad-specificity caspase inhibitor
with second-order inactivation rates; 7100, 16 000,
18 000, 180 000, 280 000 M
1
s
1
for caspases 6, 3, 7, 9,
and 8, respectively (Garcia-Calvo et al., 1998). MM.1S
cells (3 10
5
/ml) and RPMI8226 cells (1.5 10
5
/ml)
were exposed to 800 n
M VX-944 for 48 h, with or
without z-VAD-fmk (25 m
M) pretreatment for 2 h.
Neither growth inhibition nor cytotoxicity of VX-944,
as determined by trypan blue exclusion, was reduced
by pretreatment with z-VAD-fmk (Figure 3b). Cyto-
toxicity of lower concentrations of VX-944 (200 and
400 n
M) was similarly unaffected by pretreatment with
z-VAD-fmk (data not shown). Moreover, pretreatment
with z-VAD-fmk did not block induction of apoptosis
Figure 3 VX-944 induced modest cleavage of caspase 3, 8 and 9 in MM.1S cells and RPMI8226 cells, demonstrated by Western
blotting (a; control; b, cells treated with 800 n
M of VX-944 for 48 h; c, cells treated with z-VAD-fmk 25 mM for 2 h, followed by 800 nM
of VX-944 for 48 h; d, control; e, cells treated with 4 mM of As
2
O
3
for 12 h; f, cells treated with z-VAD-fmk 25 mM for 2 h, followed by
4 m
M of As
2
O
3
for 12 h). Cytotoxicity (b) and apoptosis (c) induced by VX-944 were not blocked by pretreatment with z-VAD-fmk.
Cells were treated with 800 n
M of VX-944 for 48 h, with or without z-VAD-fmk (25 mM) pretreatment. Cytotoxicity was determined by
counting viable cells by trypan-blue exclusion dye. Representative data from triplicate experiments are shown (b; MM.1S and
RPMI8226). The percentage apoptotic cells after treatment with 800 n
M of VX-944 for 48 h, with or without pretreatment of z-VAD-
fmk (25 m
M) for 2 h, was determined by flow-cytometric analysis for APO2.7 staining. Block of As
2
O
3
-induced apoptosis confirmed the
inhibiting activity of z-VAD-fmk (c)
VX-944 induces apoptosis in MM cells
K Ishitsuka et al
5891
Oncogene
by VX-944 at 48 h in both cell lines (Figure 3c).
MM.1S cells treated with arsenic trioxide (As
2
O
3
;
4 m
M) for 48 h, with or without z-VAD-fmk
pretreatment, were a control for caspase-dependent
apoptosis and the inhibitory activity of z-VAD-fmk
(Hayashi et al., 2002; McCafferty-Grad et al.,
2003).
Modulation of pro- and anti-apoptotic proteins by
VX-944 was next evaluated. MM.1S and RPMI8226
cells were treated with 800 n
M VX-944 for 12, 24, and
48 h; cell lysates were prepared as described previously.
Of bcl-2 family member proteins, Bax and Bak were
markedly upregulated in VX-944-treated cells, without
significant changes in Bcl-2, Mcl-1, XIAP, and Bad
(Figure 4a). Furthermore, translocation of mitochon-
drial proapoptotic proteins, apoptosis-inducing factor
(AIF) and endonuclease G (Endo G) to cytosolic
fractions was observed (Figure 4b).
Effect of VX-944 on MM cells cultured with exogenous
IL-6, IGF-1, and bone marrow stromal cells (BMSCs)
We next examined the effect of VX-944 on MM cells in
the presence of exogenous IL-6 and IGF-1, as well as on
MM cell growth in the BM microenvironment. Neither
IL-6 nor IGF-1 protected against VX-944-induced
growth inhibition (Figure 5a and b). Binding of
Figure 4 VX-944 enhances expression of Bax and Bak
proteins, determined by Western blotting of whole cell lysates
(a). Mitochondrial proteins AIF and Endo G were released
to cytosolic fraction after VX-944 (800 n
M) treatment in MM.1S
cells (b)
Figure 5 VX-944 overcomes protective effects of IL-6 (a), IGF-1 (b) and adherence to patient BMSCs in MM.1S cells (c, d). DNA
synthesis was determined by measuring [
3
H]thymidine incorporation during the last 8 h of 48 h cultures (*Po0.05)
VX-944 induces apoptosis in MM cells
K Ishitsuka et al
5892
Oncogene
MM.1S cells to BMSCs triggers DNA synthesis, which
was also abrogated by VX-944 (Figure 5c and d).
Importantly, VX-944 at similar concentrations did not
affect the viability of BMSCs, as determined by MTT
assay (data not shown). To further delineate the effect of
VX-944 on growth signaling in MM.1S cells, MM.1S
cells were stimulated with IL-6 (20 ng/ml) or IGF-1
(25 ng/ml) for 10 min following pretreatment with
800 n
M of VX-944 for 3, 6, and 9 h. No significant
inhibition of STAT3, p42/44 MAPK or Akt signaling
was noted in VX-944-treated MM.1S cells (data not
shown).
Combination with VX-944 and Dox or Mel augments
inhibition of growth of MM.1S cells
Sequential treatment of MM.1S cells with VX-944
followed by Dox or Mel enhanced the cytotoxicity of
each drug (Figure 6a and b). Additive effects were also
observed in MM.1S cells cultured with BMSCs derived
from MM patient (Figure 6c and d).
Discussion
In this study, we first showed that the novel IMPDH
inhibitor VX-944 directly inhibits growth of MM cell
lines. Growth inhibition of MM cell lines, including
Dox-, Mel-, and Dex-resistant lines, was observed at an
IC
50
of o600 nM by MTT assay. Importantly, our
studies further showed that up to 6.4 m
M VX-944 did not
inhibit the viability of normal PBMNCs, consistent with
a Phase I pharmacokinetic study of VX-944 in which
high-plasma concentrations were achieved without toxic
effects. These data therefore demonstrate that VX-944
effectively inhibits growth of MM cell lines, including
drug-resistant cells, at clinically achievable concentra-
tions. Neither co-culture with BMSCs, which mimics the
BM microenvironment of MM patients, nor exogenous
IL-6 and IGF-1, which are growth and survival factors
for MM cells, protected MM cells against VX-944-
induced cell death (Klein et al., 1995; Chauhan et al.,
1997; Jelinek et al., 1997; Ge and Rudikoff, 2000).
Although Gu et al. (2003) have reported that MPA
Figure 6 Sequential treatment with VX-944 followed by Dox (a, c) or Mel (b, d) demonstrates additive cytotoxicity in MM.1S cells.
MM.1S cells were treated with VX-944 for 24 h, followed by addition of combination agent. Cell growth was determined at 72 h by
MTT assay (a, b). MM.1S cells cultured with BMSCs were treated with VX-944 for 24 h, followed by the addition of combined agent.
DNA synthesis is determined by incorporation of [
3
H]thymidine during the last 8 h of 48 h culture (c, d)(*Po0.05)
VX-944 induces apoptosis in MM cells
K Ishitsuka et al
5893
Oncogene
causes downregulation of STAT3 and 5, p42/44 MAPK,
and Akt pathway in a IL-3 dependent murine hemato-
poietic cell line (Gu et al., 2003), our study did not reveal
inhibition of STAT3, p42/44 MAPK, or Akt signaling in
VX-944-treated MM.1S cells, suggesting that growth
inhibition induced by VX-944 is independent of these
pathways.
Caspases are believed to be the key executors of
apoptosis. However, recent studies suggest that caspase
activation is not the sole pathway for inducing apoptosis
or necrosis by death stimuli (Jaattela and Tschopp,
2003; Abraham and Shaham, 2004; Lockshin and
Zakeri, 2004). For example, caspase-independent cell
death in MM cells has been reported in As
2
O
3
-treated
RPMI8226 cells and patient MM cells (McCafferty-
Grad et al., 2003). Since most drugs used to treat MM
induce cell death via activation of caspases, agents such
as VX-944 that can kill MM cells via caspase-
independent pathways may be particularly useful for
combination with conventional and novel drugs to
overcome drug resistances.
During VX-944-induced apoptosis, activation of
caspases 3, 8 and 9 was only modest; caspases 6 and 7,
known mediators of caspase 3-independent apoptotic
signaling (Miyashita et al., 1998; Kagawa et al., 2001;
Pirnia et al., 2002), were not activated. Cleavage of
DFF45, which inactivates the inhibitory function of
DFF40 and related nuclear DNA fragmentation (Liu
et al., 1998), was not observed. In addition, pretreat-
ment with z-VAD did not significantly inhibit VX-944
triggered cytotoxicity or apoptosis. Therefore, VX-944
appears to induce apoptosis primarily via a caspase-
independent mechanism.
We next determined whether VX-944 induced Bax
and Bak protein expression, as well as translocation of
AIF and Endo G to the cytoplasm. The Bax/AIF/Endo
G pathway is a major caspase-independent apoptotic
cascade (Daugas et al., 2000; Ahn et al., 2004; Cande
et al., 2004; Cregan et al., 2004). In this pathway,
enhanced Bax and Bak expression induces mitochon-
drial membrane permeabilization, thereby releasing AIF
and Endo G from the mitochondria to the cytosol and
nucleus, with subsequent chromatin condensation and
cell death. Conversely, reports that apoptosis induced
via Bax/AIF/Endo G pathway can be caspase-depen-
dent show that the release of AIF and Endo G from
mitochondria is blocked by z-VAD (Arnoult et al., 2003;
Cande et al., 2004). In our study, the release of these
proapoptotic proteins is likely caspase-independent,
since caspase activation was modest.
As most conventional and novel drugs induce
apoptosis in MM cells via activation of caspases, agents
killing MM cells via caspase-independent pathways may
overcome drug resistances. Therefore, we next examined
the effect of combining VX-944 and other drugs in vitro.
The additive effect of VX-944 and Dox or Mel shown in
our study indicates that VX-944 can be combined with
these drugs to enhance their efficacy. Sequential treat-
ment with VX-944, followed by Dox or Mel, holds great
promise, since these combinations inhibited the growth
of MM cells even in the presence of BMSCs.
In conclusion, we have shown that the novel IMPDH
inhibitor VX-944 appears to induce apoptosis mainly
via a caspase-independent, Bax/AIF/Endo G pathway
in MM cells. VX-944 induces cell death in drug-resistant
MM cells and overcomes the growth-promoting activity
of IL-6, IGF-1 and BMSCs. These results suggest that
VX-944, used either alone or in combination with other
drugs, represents a promising novel targeted approach
to enhance MM cell cytotoxicity and improve patient
outcome in MM.
Materials and methods
Cells
Dex-sensitive MM.1S and Dex-resistant MM.1R human MM
cell lines were kindly provided by Dr Steven Rosen (North-
western University, Chicago, IL, USA). RPMI 8226-Dox40
(Dox-resistant) and RPMI 8226-LR5 (Mel-resistant) human
MM cell lines were kindly provided by Dr William Dalton
(Moffit Cancer Center, Tampa, FL, USA). RPMI-8226 and
U266 cells were obtained from the American Type Culture
Collection (Rockville, MD, USA). Fresh PBMNCs obtained
from three healthy subjects after informed consent were
separated from heparinized peripheral blood by Ficoll-
Hipaque density sedimentation. Cells were cultured at 371C
in RPMI 1640 containing 10% fetal bovine serum (FBS;
Sigma, St Louis, MO, USA), 2 m
ML-glutamine, 100 U/ml
penicillin, and 100 mg/ml streptomycin (Gibco, Grand Island,
NY, USA).
MNCs in BM specimens, obtained from patients with MM
after informed consent, were separated by Ficoll-Hipaque
density sedimentation and used to establish long-term BMSC
cultures, as described (Uchiyama et al., 1993; Hideshima et al.,
2001).
Reagents
VX-944 (Vertex Pharmaceuticals, Cambridge, MA, USA) was
dissolved in dimethyl sulfoxide (DMSO) at 10 m
M stock
solution and stocked at 201C. Guanosine and Dox (Sigma,
St Louis, MO, USA) were dissolved in sterile water at 1 and
3.45 m
M, respectively. Mel (Sigma, St Louis, MO, USA) was
dissolved in DMSO at 20 m
M. Recombinant human IL-6 and
IGF-1 (R&D Systems, Minneapolis, MN, USA) were recon-
stituted with sterile PBS containing 0.1% FBS and 10 m
M
acetic acid containing 0.1% FBS, respectively. Pan-caspase
inhibitor z-VAD-fmk (Bachem, Bubendorf, Switzerland) was
dissolved in methanol.
Cellular proliferation and DNA synthesis assay
Colorimetric assays were performed to evaluate drug activity.
MM cell lines and MM patient BMSCs were cultured in 96-
well culture plates with VX-944 in 100 ml of media for 48 h,
pulsed with 10 ml of 5 mg/ml 3-(4,5-dimethylthiazol-2-yl)-2,5-
diphenyl tetrasodium bromide (MTT; Sigma, St Louis, MO,
USA) to each well for 4 h, followed by 100 ml isopropanol
containing 0.04
N HCl. Absorbance readings at a wavelength
of 570 nm and reference wave length of 630 nm were taken on a
spectrophotometer (Molecular Devices Corp., Sunnyvale, CA,
USA).
DNA synthesis was measured by [
3
H]thymidine uptake, as
described previously (Hideshima et al., 2000). Cells in 96-well
culture plates were pulsed with 0.5 mCi/well of [
3
H]thymidine
VX-944 induces apoptosis in MM cells
K Ishitsuka et al
5894
Oncogene
(Perkin-Elmer, Boston, MA, USA) during the last 8 h of
culture, harvested onto glass filters with an automatic cell
harvester (Cambridge Technology, Cambridge, MA, USA),
and counted using the LKB Betaplate scintillation counter
(Wallac, Gaithersburg, MD, USA). All experiments were
performed in triplicate.
Detection of cytotoxicity and apoptosis
Cytotoxicity was determined by trypan blue exclusion assay
(Sigma, St Louis, MO, USA). Viability was expressed as
percent viable cells. Apoptosis in VX-944-treated cells was
assesed by annexin V and PI staining, TUNEL assay, and
APO 2.7 staining. For annexin V and PI staining, cells were
suspended in binding buffer (10 mmol/l HEPES, pH 7.4,
140 mmol/l NaCl, 2.5 mmol/l CaCl
2
), followed by incubation
with annexin V-FITC (MBL, Nagoya, Japan) and PI (MBL,
Nagoya, Japan) for 5 min at room temperature. To detect
DNA strand breaks, the TUNEL assay was performed using a
commercial kit (MBL, Nagoya, Japan). In brief, cells were
fixed and permeabilized by 4% paraformaldehyde and 70%
ethanol, followed by incubation with a mixture of FITC-dUTP
and TdT for 1 h at 371C. For detection of mitochondrial
membrane protein 7A6 expressed in apoptotic cells, cells were
incubated with APO 2.7 reagent (Immunotech, Marseille,
France) for 20 min. Expression of Annexin V and PI, TUNEL,
and APO 2.7 was determined using an EPICS XL flow
cytometer.
Western blotting
MM cells were harvested, washed twice with ice-cold PBS, and
lysed in lysis buffer: 50 m
M Tris-HCl (pH 7.4), 150 mM NaCl,
1% NP-40, 5 m
M EDTA, 5 mM NaF, 2 mM Na
3
V
4
,1mM
PMSF, 5 mg/ml leupeptine, and 5 mg/ml aprotinin after VX-944
treatment. Subcellular proteins were extracted from 2 10
7
viable cells using mitochondria isolation kit (Pierce, Rockford,
IL, USA). Whole-cell lysates or fractionated proteins were
subjected to SDS–PAGE, transferred to nitrocellulose mem-
brane, and immunoblotted with anti-caspase-3, caspase-6,
caspase-7, caspase-8, caspase-9, DFF45/35, Bax, Bak, XIAP,
AIF, phospho-Akt, and Akt (Cell Signaling, Beverly, MA,
USA); phospho-p42/44 MAPK, phospho-STAT3, bcl-2, and
Mcl-1 (Santa Cruz Biotechnology, Santa Cruz, CA, USA); and
Endo G (Axxora, San Diego, CA, USA) antibodies. Immuno-
blotting with anti-alpha-tubulin Ab (Sigma, St Louis, MO,
USA) and anti-voltage-dependent anion channel (VDAC) Ab
(Calbiochem, San Diego, CA, USA) confirmed equivalent
protein loading.
Effect of IL-6, IGF-1, and BMSCs on VX-944-induced growth
inhibition
MM.1S cells were cultured for 48 h with VX-944, in the
presence or absence of IL-6 or IGF-1. To evaluate growth
stimulation in MM cells adherent to BMSCs, MM.1S cells
were cultured in BMSC-coated 96-well plates for 48 h, in the
presence or absence of VX-944. DNA synthesis in these
cultures was measured by [
3
H]thymidine uptake.
Combination with other drugs
MM.1S cells were cultured with VX-944 for 24 h, followed by
addition of either Dox or Mel. Cellular growth was determined
by MTT assay after 72 h treatment. Cells were also incubated
with VX-944 in BMSC-coated 96-well plates, followed by
addition of either of Dox or Mel at 24 h. DNA synthesis was
measured by the addition of [
3
H]thymidine uptake for the last
8 h of the 48 h cultures.
Acknowledgements
We acknowledge K Lin, R Hoover, Y Yao, M Harding, J
Thomson, M Partridge and C Sorensen at Vertex for critical
reading of the manuscript or advice during the course of these
studies. This study is supported by National Institutes of
Health Grants Specialized Programs of Research Excellence
(SPORE) IP50 CA10070-01, PO-1 78378, and RO-1 CA
50947; the Doris Duke Distinguished Clinical Research
Scientist Award (KCA); the Multiple Myeloma Research
Foundation (TH); and the Cure for Myeloma Research Fund
(KCA).
References
Abraham MC and Shaham S. (2004). Trends Cell Biol., 14,
184–193.
Ahluwalia GS, Jayaram HN, Plowman JP, Cooney DA
and Johns DG. (1984). Biochem. Pharmacol., 33, 1195–
1203.
Ahn HJ, Kim YS, Kim JU, Han SM, Shin JW and Yang HO.
(2004). J. Cell. Biochem., 91, 1043–1052.
Arnoult D, Gaume B, Karbowski M, Sharpe JC, Cecconi F
and Youle RJ. (2003). EMBO J., 22, 4385–4399.
Cande C, Vahsen N, Garrido and Kroemer G. (2004). Cell
Death Differ., 11, 591–595.
Chauhan D, Kharbanda S, Ogata A, Urashima M, Teoh G,
Robertson M, Kufe DW and Anderson KC. (1997). Blood,
89, 227–234.
Cregan SP, Dawson VL and Slack RS. (2004). Oncogene, 23,
2785–2796.
Daugas E, Susin SA, Zamzami N, Ferri KF, Irinopoulou T,
Larochette N, Prevost MC, Leber B, Andrews D, Penninger
J and Kroemer G. (2000). FASEB J., 14, 729–739.
Eugui EM and Almquist SJ. (1990). Proc. Natl. Acad. Sci.
USA, 87, 1305–1309.
Eugui EM, Almquist SJ, Muller CD and Allison AC. (1991).
Scand. J. Immunol., 33, 161–173.
Garcia-Calvo M, Peterson EP, Leiting B, Ruel R, Nicholson
DW and Thornberry NA. (1998). J. Biol. Chem., 273, 32608–
32613.
Ge NL and Rudikoff S. (2000). Blood, 96, 2856–2861.
Gu JJ, Gathy K, Santiago L, Chen E, Huang M, Graves LM
and Mitchell BS. (2003). Blood, 101, 4958–4965.
Hayashi T, Hideshima T, Akiyama M, Richardson P,
Schlossman RL, Chauhan D, Munshi NC, Waxman and
Anderson KC. (2002). Mol. Cancer Ther., 1, 851–860.
Hideshima T, Chauhan D, Schlossman R, Richardson and
Anderson KC. (2001). Oncogene, 20, 4519–4527.
Hideshima T, Chauhan D, Shima Y, Raje N, Davies FE, Tai
YT, Treon SP, Lin B, Schlossman RL, Richardson P, Muller
G, Stirling DI and Anderson KC. (2000). Blood, 96,
2943–2950.
Hunakova L, Bies J, Sedlak J, Duraj J, Jakubikova J,
Takacsova X, Novotny L and Chorvath B. (2000).
Neoplasma, 47, 274–279.
Jaattela M and Tschopp J. (2003). Nat. Immunol., 4, 416–423.
Jain J, Almquist SJ, Shlyakhter and Harding MW. (2001).
J. Pharm Sci., 90, 625–637.
Jelinek DF, Witzig TE and Arendt BK. (1997). J. Immunol.,
159, 487–496.
VX-944 induces apoptosis in MM cells
K Ishitsuka et al
5895
Oncogene
Kagawa S, Gu J, Honda T, McDonnell TJ, Swisher SG, Roth
JA and Fang B. (2001). Clin. Cancer Res., 7, 1474–1480.
Klein B, Zhang XG, Lu ZY and Bataille R. (1995). Blood, 85,
863–872.
Knudtzon S and Nissen NI. (1972). Cancer Chemother. Rep.,
56, 221–227.
Laliberte J, Yee A, Xiong Y and Mitchell BS. (1998). Blood,
91, 2896–2904.
Liu X, Li P, Widlak P, Zou H, Luo X, Garrard WT and Wang
X. (1998). Proc. Natl. Acad. Sci. USA, 95, 8461–8466.
Lockshin RA and Zakeri Z. (2004). Oncogene, 23, 2766–2773.
McCafferty-Grad J, Bahlis NJ, Krett N, Aguilar TM, Reis I,
Lee KP and Boise LH. (2003). Mol. Cancer Ther., 2,
1155–1164.
McLean JE, Hamaguchi N, Belenky P, Mortimer SE,
Stanton M and Hedstrom L. (2004). Biochem. J., 379,
243–251.
Messina E, Gazzaniga P, Micheli V, Guaglianone MR,
Barbato S, Morrone S, Frati L, Agliano AM and
Giacomello A. (2004). Int. J. Cancer, 108, 812–817.
Miyashita T, Nagao K, Krajewski S, Salvesen GS, Reed JC,
Inoue T and Yamada M. (1998). Cell Death Differ., 5,
1034–1041.
Nagai M, Natsumeda Y, Konno Y, Hoffman R, Irino S and
Weber G. (1991). Cancer Res., 51, 3886–3890.
Pirnia F, Schneider E, Betticher DC and Borner MM. (2002).
Cell Death Differ., 9, 905–914.
Richardson P, Schlossman R, Jagannath S, Alsina M, Desikan
R, Blood E, Weller E, Mitsiades C, Hideshima T, Davies F,
Doss D, Freeman A, Bosch J, Patin J, Knight R, Zeldis J,
Dalton W and Anderson K. (2004). Mayo Clin. Proc., 79,
875–882.
Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath
S, Irwin D, Rajkumar SV, Srkalovic G, Alsina M, Alexanian
R, Siegel D, Orlowski RZ, Kuter D, Limentani SA, Lee S,
Hideshima T, Esseltine DL, Kauffman M, Adams J,
Schenkein DP and Anderson KC. (2003). N. Engl.
J. Med., 348, 2609–2617.
Richardson PG, Schlossman RL, Weller E, Hideshima T,
Mitsiades C, Davies F, LeBlanc R, Catley LP, Doss D, Kelly
K, McKenney M, Mechlowicz J, Freeman A, Deocampo R,
Rich R, Ryoo JJ, Chauhan D, Balinski K, Zeldis J and
Anderson KC. (2002). Blood, 100, 3063–3067.
Senda M and Natsumeda Y. (1994). Life Sci., 54, 1917–1926.
Sweeney MJ, Gerzon K, Harris PN, Holmes RE, Poore GA
and Williams RH. (1972). Cancer Res., 32, 1795–1802.
Szekeres T, Sedlak J and Novotny L. (2002). Curr. Med.
Chem., 9, 759–764.
Takebe N, Cheng X, Wu S, Bauer K, Goloubeva OG, Fenton
RG, Heyman M, Rapoport AP, Badros A, Shaughnessy J,
Ross D, Meisenberg B and Tricot G. (2004). Clin. Cancer
Res., 10, 8301–8308.
Turka LA, Dayton J, Sinclair G, Thompson CB and Mitchell
BS. (1991). J. Clin. Invest, 87, 940–948.
Uchiyama H, Barut BA, Mohrbacher AF, Chauhan D and
Anderson KC. (1993). Blood, 82, 3712–3720.
Wright DG, Boosalis MS, Waraska K, Oshry LJ, Weintraub
LR and Vosburgh E. (1996). Anticancer Res., 16, 3349–3351.
Yalowitz JA and Jayaram HN. (2002). Curr. Med. Chem, 9,
749–758.
Yalowitz JA, Pankiewicz K, Patterson SE and Jayaram HN.
(2002). Cancer Lett., 181, 31–38.
Zimmermann AG, Gu JJ, Laliberte J and Mitchell BS. (1998).
Prog. Nucleic Acid Res. Mol. Biol., 61, 181–209.
VX-944 induces apoptosis in MM cells
K Ishitsuka et al
5896
Oncogene
... A related compound, VX-944, was found to have broad anti-cancer properties in vitro and was investigated further [139] (Figure 4E). VX-944 is an orally bioavailable, small-molecule, noncompetitive inhibitor of both human IMPDH1 and IMPDH2 [140]. VX-944 was developed using a structure-based drug design program and displayed a novel mode of interaction with IMPDH. ...
... The mice treated with VX-944 had significantly longer median survival time compared to those treated with standard therapy. VX-944 suppressed proliferation of multiple myeloma cell lines, including drug-resistant cells [140], and several human cancer cell lines including colon, breast, lung, pancreatic, melanoma, and prostate [142]. A related compound, VX-944, was found to have broad anti-cancer properties in vitro and was investigated further [139] (Figure 4E). ...
... A related compound, VX-944, was found to have broad anti-cancer properties in vitro and was investigated further [139] (Figure 4E). VX-944 is an orally bioavailable, small-molecule, non-competitive inhibitor of both human IMPDH1 and IMPDH2 [140]. VX-944 was developed using a structure-based drug design program and displayed a novel mode of interaction with IMPDH. ...
Article
Full-text available
The purine nucleotides ATP and GTP are essential precursors to DNA and RNA synthesis and fundamental for energy metabolism. Although de novo purine nucleotide biosynthesis is increased in highly proliferating cells, such as malignant tumors, it is not clear if this is merely a secondary manifestation of increased cell proliferation. Suggestive of a direct causative effect includes evidence that, in some cancer types, the rate-limiting enzyme in de novo GTP biosynthesis, inosine monophosphate dehydrogenase (IMPDH), is upregulated and that the IMPDH inhibitor, mycophenolic acid (MPA), possesses anti-tumor activity. However, historically, enthusiasm for employing IMPDH inhibitors in cancer treatment has been mitigated by their adverse effects at high treatment doses and variable response. Recent advances in our understanding of the mechanistic role of IMPDH in tumorigenesis and cancer progression, as well as the development of IMPDH inhibitors with selective actions on GTP synthesis, have prompted a reappraisal of targeting this enzyme for anti-cancer treatment. In this review, we summarize the history of IMPDH inhibitors, the development of new inhibitors as anti-cancer drugs, and future directions and strategies to overcome existing challenges.
... Apoptosis triggered by these inhibitors occurs through multiple pathways depending on the type of compound and the used tumoural model. Figure 5 summarizes the apoptotic pathways triggered by the best known IMPDH inhibitors [136,137,[162][163][164][165][166][167][168][169][170]. In all the experiments, apoptotic mechanisms were reversed by the simultaneous addition of guanosine, that, through the salvage pathway, circumvents the IMPDH inhibition by restoration of the guanylate pool. ...
... In all the experiments, apoptotic mechanisms were reversed by the simultaneous addition of guanosine, that, through the salvage pathway, circumvents the IMPDH inhibition by restoration of the guanylate pool. [136,162,163]; (3): upregulation of p53 with (a) downregulation of Bcl-2 and Bcl-xL, with consequent inhibition of p27 and survivin, cytochrome c (Cyt c) release, and activation of caspase-9, caspase-3 and polyADP-ribose polymerase (PARP; intrinsic apoptotic pathway) [159][160][161]164,165], (b) upregulation of PUMA and BIM with consequent SMAC/DIABLO release from mitochondria, inhibition of Inhibitor of Apoptosis (IAPs) (a caspase-3 inhibitor) with activation of caspase-3 [164], and (c) activation of caspase-2 with cleavage of Bid into truncated Bid (t-Bid) and AIF/Endo G release from mitochondria [164]; (4): synergistic effect of IMPDH inhibitors with TRAIL through binding with death receptors (DR4 and DR5) which recruit initiator caspase-8 via the adaptor protein FADD. Activated caspase-8 stimulates apoptosis via two parallel cascades: direct cleavage and activation of caspase-3, or cleavage of Bid into t-Bid which translocates to mitochondria, inducing cytochrome c release, with sequential activation of caspase-9 and -3 (extrinsic apoptotic pathway) [164]. ...
Article
Full-text available
The enzymes of both de novo and salvage pathways for purine nucleotide synthesis are regulated to meet the demand of nucleic acid precursors during proliferation. Among them, the salvage pathway enzymes seem to play the key role in replenishing the purine pool in dividing and tumour cells that require a greater amount of nucleotides. An imbalance in the purine pools is fundamental not only for preventing cell proliferation, but also, in many cases, to promote apoptosis. It is known that tumour cells harbour several mutations that might lead to defective apoptosis-inducing pathways, and this is probably at the basis of the initial expansion of the population of neoplastic cells. Therefore, knowledge of the molecular mechanisms that lead to apoptosis of tumoural cells is key to predicting the possible success of a drug treatment and planning more effective and focused therapies. In this review, we describe how the modulation of enzymes involved in purine metabolism in tumour cells may affect the apoptotic programme. The enzymes discussed are: ectosolic and cytosolic 5′-nucleotidases, purine nucleoside phosphorylase, adenosine deaminase, hypoxanthine-guanine phosphoribosyltransferase, and inosine-5′-monophosphate dehydrogenase, as well as recently described enzymes particularly expressed in tumour cells, such as deoxynucleoside triphosphate triphosphohydrolase and 7,8-dihydro-8-oxoguanine triphosphatase.
... EMT is a hallmark of tumor invasion and metastasis, which can alter the adhesion of epithelial cancer cells, promoting them to invade and migrate to other distant sites (26)(27)(28). During EMT, the repression of genes encoding epithelial cell junction proteins is accompanied by the activation of genes promoting mesenchymal adhesion (29), and the 'Cadherin switch' refers to the process by which the downregulation of E-cadherin expression levels are Figure 6. ...
Article
Inosine 5'-monophosphate dehydrogenase type II (IMPDH2) is an important enzyme involved in the biosynthesis of guanine nucleotides. Therefore, the present study aimed to investigate the potential and molecular mechanism of IMPDH2 in non-small cell lung cancer (NSCLC). Reverse transcription-quantitative PCR and immunohistochemistry were used to detect IMPDH2 expression levels in NSCLC tissues and cells. A Cell Counting Kit-8 assay, colony formation assay, flow cytometry, wound healing, Transwell assay, western blotting and immunofluorescence analyses were utilized to identify the effects of upregulated IMPDH2 levels on NSCLC cells. The expression levels of IMPDH2 have been discovered to be upregulated in several types of human cancer; however, the biological and clinical value of IMPDH2 in NSCLC remains unclear. The results of the present study revealed that the expression levels of IMPDH2 were significantly upregulated in NSCLC tissues. Furthermore, the genetic knockdown of IMPDH2 significantly hindered the proliferation, apoptosis, invasion, migration and epithelial-mesenchymal transition of NSCLC cells, whereas the overexpression of IMPDH2 achieved the opposite results. In addition, the results of the present study demonstrated that the inhibition of IMPDH2 inhibited the Wnt/β-catenin signaling pathway by decreasing the expression levels of Wnt3a and β-catenin, while increasing the expression levels of phosphorylated glycogen synthase kinase-3β in NSCLC cells. These findings of the present study indicated that IMPDH2 may promote NSCLC progression by activating the Wnt/β-catenin signaling pathway, which suggested that IMPDH2 may be a novel therapeutic target for patients with NSCLC.
... Other distinct MPA analogs have been developed which were structurally distinct from MPA and the nucleoside analogs [91]. Of these, compound VX-944, developed by Vertex pharmaceuticals using a structure-based drug design approach, strongly inhibited hIMPDH1 and hIMPH2 (K i = 6 and 10 nM, respectively) [92] and showed broad anti-cancer properties in vitro [42,93]. Unlike Tiazofurin, VX-944 does not require intracellular activation, hence circumventing one of the mechanisms of resistance to Tiazofurin. ...
Article
Dehydrogenases are oxidoreductase enzymes that play a variety of fundamental functions in the living organisms and have primary roles in pathogen survival and infection processes as well as in cancer development. We review here a sub-set of NAD-dependent dehydrogenases involved in human diseases and the recent advancements in drug development targeting pathogen-associated NAD-dependent dehydrogenases. We focus also on the molecular aspects of the inhibition process listing the structures of the most relevant molecules targeting this enzyme family. Our aim is to review the most impacting findings regarding the discovery of novel inhibitory compounds targeting the selected NAD-dependent dehydrogenases involved in cancer and infectious diseases.
... AIF is an oxidoreductase located in the mitochondrial membrane space and is considered a scavenger of reactive oxygen species (ROS). It has the dual role of maintaining normal mitochondria and inducing cell death [35,36]. In the process of parthanatos, AIF acts as an important mediator in the production of chromosome condensation and large fragmentation of the DNA in the nucleus. ...
Article
Full-text available
Cerebral ischemia–reperfusion injury is a complex pathophysiological process. Poly(ADP-ribose) (PAR) polymerase-1 (PARP-1)/apoptosis-inducing factor (AIF) signaling pathway-mediated apoptosis is one of the non-caspase-dependent cell death programs that are widely present in neurological diseases such as stroke. In our study, we aimed to conduct further research on the effects of Gualou Guizhi decoction (GLGZD) on the PARP-1/AIF signaling pathway in cell apoptosis after ischemia–reperfusion injury caused by middle cerebral artery occlusion (MCAO). The results showed that GLGZD administration for 7 days significantly ameliorated MCAO-induced neurological damage, limb paralysis and the pathological state of the ischemic cortex. GLGZD exerted its effects by significantly reducing the volume of ischemic cerebral infarction, increasing the number of Nissl-positive cells, and reducing neuronal apoptosis. Furthermore, Western blot analysis showed that GLGZD significantly inhibited the total protein expression of PARP-1, PAR, AIF and endonuclease G (Endo G) in the ischemic cortex and significantly increased the total protein expression of heat-shock protein 70 (Hsp70). On the one hand, the expression of PARP-1, AIF and Endo G protein in the nucleus significantly decreased while the expression of PAR nucleoprotein significantly upregulated. On the other hand, compared with the MCAO model group, the GLGZD-treated group showed a significantly reduced protein expression of PAR in mitochondria and significantly increased protein expression of mitochondrial AIF and Endo G. It was concluded that GLGZD had good therapeutic effects in MCAO model rats. These effects were closely related to GLGZD-mediated inhibition of ischemia-induced neuronal apoptosis by regulation of protein expression and translocation in the PARP-1/AIF signaling pathway.
... AVN944 is a highly selective, non-nucleoside, specific, uncompetitive IMPDH inhibitor, which was developed by Vertex (VX-944) and licensed by Avalon Pharmaceuticals (AVN944) [16]. AVN944 has been demonstrated to be well-tolerated by humans, and is currently under clinical trials on patients with hematological malignances and, in combination with gemcitabine, on patients with pancreatic cancer [17]. ...
Article
Full-text available
Foot-and-mouth disease virus (FMDV) is an important pathogen that affects livestock breeding and causes huge economic losses worldwide. Currently, the development of antiviral agents to combat FMDV infection at the early stages is being explored. As viral replication critically depends on the host for nucleoside supply, host enzymes involved in nucleotides biosynthesis may represent potential targets for the development of antiviral agents. In the present study, the effects of IMP dehydrogenase (AVN-944 and mycophenolate mofetil) and dihydroorotate dehydrogenase (teriflunomide) inhibitors were evaluated both in vitro and in vivo. The results revealed that these compounds were effective in suppressing FMDV (O/MY98/BY/2010 and A/GD/MM/2013) infection. With regard to the antiviral mechanism, time-of-addition experiments revealed that these compounds were effective when added at the early stages of viral lifecycle (0-8 h post infection). However, exogenous guanosine/uridine eliminated the antiviral activity of these compounds. Importantly, treatment AVN-944 and teriflunomide significantly improved the survival of mice that were subcutaneously treated with FMDV. Together, the results of the present study indicate the broad-spectrum activities of anti-FMDV agents targeting IMP dehydrogenase or dihydroorotate dehydrogenase, which could be useful in developing strategies to prevent FMD.
Article
Four novel Gelsemium elegans cyclic peptides (GEPs) were isolated in an anti‐human cervical carcinoma activity tracking method, and amino acid sequence identified. GEP‐1 cyclic‐(Trp‐Leu‐His‐Val)‐peptide inhibited HeLa cell proliferation in a dose‐ and time‐dependent manner. GEP‐1 induced intracellular reactive oxygen species (ROS) over‐production, and induced HeLa cells apoptosis in a Caspase‐dependent manner. GEP‐1 also induced collapse of mitochondrial membrane potential, and promoted the mitochondria releases of Cyt c, AIF and Endo G in HeLa cells. Furthermore, GEP‐1 triggered extrinsic death receptor‐dependent pathway, which was characterized by activating Fas and FADD. Notably, the GEP‐1 is a potential anti‐human cervical carcinoma peptide.
Article
Liver injury could be caused by a variety of causes, including alcohol, drug poisoning, autoimmune overreaction, etc. In the period of liver injury, hepatic stellate cells (HSCs) will be activated and produce excessive extracellular matrix (ECM). If injury cannot be suppressed, liver injury will develop into fibrosis, even cirrhosis and liver cancer. It is reported that some monomer components extracted from traditional Chinese medicine have better effects on protecting liver. Emodin, an anthraquinone compound extracted from the traditional Chinese medicine RHEI RADIX ET RHIZOMA, has anti-inflammatory, antioxidant, liver protection and anti-cancer effects, and can prevent liver injury induced by a variety of factors. By searching literatures related to the liver protection of emodin in PUBMED, SINOMED, EBM and CNKI databases, it was found that emodin could inhibit the production and promote the secretion of bile acids, and have a protective effect on intrahepatic cholestasis. Also, emodin reduce collagen synthesis and anti-hepatic fibrosis by inhibiting oxidative stress, TGF-β/Smad pathway and HSCs proliferation, and promoting apoptosis of HSCs. Emodin can also regulate lipid metabolism and regulate the synthesis and oxidation of lipids and cholesterol to protect the nonalcoholic fatty liver. Besides, emodin can induce the apoptosis of hepatocellular carcinoma cells by acting on the death receptor pathway and mitochondrial apoptosis pathway, thus inhibiting the development of hepatocellular carcinoma. Moreover, emodin can modulate immunity and improve immune rejection in liver transplantation animals. In conclusion, emodin has a good effect on liver protection, but further experimental data are needed to verify it.
Article
Bisdemethoxycurcumin (BDMC) is one of three curcuminoids extracted from turmeric. Unlike the dominant ingredient curcumin with some intensive investigations, BDMC was recently reported to possess potent anti-tumor, anti-inflammatory, anti-atherosclerosis, anti-obesity and anti-ageing effects. Considering its pharmacological effects in inflammation, atherosclerosis and obesity, this study was designed to examine if BDMC displays cardioprotective property. In this study, staurosporine (STS) was used to establish cardiomyocyte injury model. Our data revealed that BDMC significantly inhibited myocardial apoptosis, improved cell survival, reduced caspase-3 activity and diminished reactive oxygen species (ROS) production. BDMC enhanced phosphorylation of protein kinase B (AKT) and extracellular signal-regulated kinase (ERK) and up-regulated the expression of HO-1. Inhibition of HO-1 activity by using tin-protoporphyrin (SnPPIX) can restrain the anti-apoptotic effect of BDMC. Furthermore, translocation of Nrf2 from cytoplasm to nucleus was ablated by LY294002, although only partially by PD98059. Up-regulation of HO-1 was weakly suppressed by PD98059 but strongly inhibited by LY294002. Unlike PD98059, LY294002 negated the protective effect of BDMC. These findings indicated that BDMC possessed favorable cardioprotection in a Nrf2 / HO-1-dependent manner. Activation of Nrf2 /HO-1 mainly depended on PI3K / AKT but not MEK/ERK signaling.
Article
FF-10501 is a novel inhibitor of inosine monophosphate dehydrogenase (IMPDH). Clinical trials of FF-10501 for myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are currently being conducted in the United States. Although it has been shown that FF-10501 induces apoptosis in hematological malignant cells, the intracellular mechanisms of this effect have not been characterized. We conducted an in vitro study to elucidate the mechanisms of FF-10501-induced cell death using 12 hematological malignant cell lines derived from myeloid and lymphoid malignancies. FF-10501 suppressed the growth of each cell line in a dose-dependent manner. However, the clinically relevant dose (40 μM) of FF-10501 induced cell death in three cell lines (MOLM-13, OCI-AML3, and MOLT-3). Investigation of the cell death mechanism suggested that FF-10501 induces both apoptotic and necrotic cell death. FF-10501-induced apoptosis was mediated by caspase-8 activation followed by activation of the mitochondrial pathway in MOLM-13 and MOLT-3 cells. FF-10501 induced necrotic cell death via endoplasmic reticulum stress in OCI-AML3 cells. The present study is the first to identify intracellular pathways involved in FF-10501-induced cell death.
Article
Fas belongs to the family of type-1 membrane proteins that transduce apoptotic signals. In the present studies, we characterized signaling during Fas-induced apoptosis in RPMI-8226 and IM-9 multiple myeloma (MM) derived cell lines as well as patient plasma cell leukemia cells. Treatment with anti-Fas (7C11) monoclonal antibody (MoAb) induced apoptosis, evidenced by internucleosomal DNA fragmentation and propidium iodide staining, and was associated with increased expression of c-jun early response gene. We also show that anti-Fas MoAb treatment is associated with activation of stress-activated protein kinase (SAPK) and p38 mitogen-activated protein kinase (MAPK); however, no detectable increase in extracellular signal-regulated kinases (ERK1 and ERK2) activity was observed. Because interleukin-6 (IL-6) is a growth factor for MM cells and inhibits apoptosis induced by dexamethasone and serum starvation, we examined whether IL-6 affects anti-Fas MoAb-induced apoptosis and activation of SAPK or p38 MAPK in MM cells. Culture of MM cells with IL-6 before treatment with anti-Fas MoAb significantly reduced both DNA fragmentation and activation of SAPK, without altering induction of p38 MAPK activity. These results therefore suggest that anti-Fas MoAb-induced apoptosis in MM cells is associated with activation of SAPK, and that IL-6 may both inhibit apoptosis and modulate SAPK activity.
Article
Previous studies show that human myeloma-derived cell lines specifically adhere to fibronectin (FN) through very late antigen-4 (VLA-4; alpha 4 beta 1 integrin complex) and RGD-peptide mechanisms, which may contribute to the localization of tumor cells in bone marrow (BM). In these studies, we characterized the adhesion of myeloma- derived cell lines to both normal and myeloma BM stromal cells (BMSCs) and the effect of adhesion on DNA synthesis. Because interleukin-6 (IL- 6) plays an important role in the pathogenesis of multiple myeloma, we also examined the effects of tumor cell adhesion on IL-6 secretion by BMSCs. In 51chromium binding assays, the U266, ARH-77, and IM-9 cell lines showed 52% +/- 12%, 55% +/- 6%, and 47% +/- 7% specific adherence, respectively, to normal BMSCs and 74% +/- 4%, 60% +/- 3%, and 61% +/- 6% specific adherence, respectively, to myeloma BMSCs. In contrast, only 12% to 13% specific binding of HS-Sultan cells to BMSCs was noted. The binding of myeloma cells to BMSCs was partially blocked with anti-beta 1 monoclonal antibody (MoAb), anti-beta 2 integrin MoAb, and excess RGD peptide, suggesting multiple mechanisms for the adhesion of myeloma cell lines to BMSCs. Binding of cell lines to FN or myeloma BMSCs did not affect cell line proliferation; however, adhesion of myeloma cell lines to normal BMSCs decreased DNA synthesis, ie, stimulation indices are 0.1 +/- 0.04, 0.2 +/- 0.1, 0.2 +/- 0.07, and 0.1 +/- 0.06 for the adherent non-IL-6-dependent U266, ARH-77, HS- Sultan, and IM-9 cells, respectively (n = 5, P < .01). In contrast, adherence of IL-6-dependent B9 cells increased their proliferation (stimulation index, 3.2 +/- 0.7). Significant (twofold to eightfold) increases in IL-6 secretion were evident in cell line-adherent (> or = 12 hours) normal and myeloma BMSC cultures. Paraformaldehyde fixation of BMSCs before adhesion completely abrogated IL-6 secretion, suggesting that IL-6 secretion was triggered in BMSCs rather than in cell lines. Partial blocking of cell line adhesion to BMSCs, using anti- beta 1 integrin and anti-beta 2 integrin MoAbs and RGD peptide, also partially blocked the triggering of IL-6 secretion by BMSCs. When cell lines were placed in Transwell inserts and then cultured with either normal or myeloma BMSCs, permitting juxtaposition without cell to cell contact between myeloma cell lines and BMSCs, no increase in IL-6 secretion was observed.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Multiple myeloma (MM) is an invariably fatal disease that accounts for approximately 1% to 2% of all human cancers. Surprisingly little is known about the cellular pathways contributing to growth of these tumors. Although the cytokine interleukin-6 has been suggested to be the major stimulus for myeloma cell growth, the role of a second potential growth factor, insulin-like growth factor I (IGF-I), has been less clearly defined. The IGF-I signaling cascade in 8 MM cell lines was examined. In 7 of these, the IGF-I receptor (IGF-IR) was expressed and autophosphorylated in response to ligand. Downstream of IGF-IR, insulin receptor substrate 1 was phosphorylated, leading to the activation of phosphatidylinositol-3′-kinase (PI-3K). PI-3K, in turn, regulated 2 distinct pathways. The first included Akt and Bad, leading to an inhibition of apoptosis; the second included the mitogen-activated protein kinase (MAPK), resulting in proliferation. Biologic relevance of this pathway was demonstrated because in vitro IGF-I induced both an antiapoptotic and a proliferative effect. Importantly, in vivo administration of IGF-I in SCID mice inoculated with the OPM-2 line led to approximately twice the growth rate of tumor cells as in controls. These results suggest that IGF-I activates at least 2 pathways effecting myeloma cell growth and contributes significantly to expansion of these cells in vivo.
Article
Although thalidomide (Thal) was initially used to treat multiple myeloma (MM) because of its known antiangiogenic effects, the mechanism of its anti-MM activity is unclear. These studies demonstrate clinical activity of Thal against MM that is refractory to conventional therapy and delineate mechanisms of anti-tumor activity of Thal and its potent analogs (immunomodulatory drugs [IMiDs]). Importantly, these agents act directly, by inducing apoptosis or G1 growth arrest, in MM cell lines and in patient MM cells that are resistant to melphalan, doxorubicin, and dexamethasone (Dex). Moreover, Thal and the IMiDs enhance the anti-MM activity of Dex and, conversely, are inhibited by interleukin 6. As for Dex, apoptotic signaling triggered by Thal and the IMiDs is associated with activation of related adhesion focal tyrosine kinase. These studies establish the framework for the development and testing of Thal and the IMiDs in a new treatment paradigm to target both the tumor cell and the microenvironment, overcome classical drug resistance, and achieve improved outcome in this presently incurable disease.
Article
Depletion of guanine nucleotide pools after inhibition of inosine monophosphate dehydrogenase (IMPDH) potently inhibits DNA synthesis by arresting cells in G1 and has been shown to induce the differentiation of cultured myeloid and erythroid cell lines, as well as chronic granulocytic leukemic cells after blast transformation. Inhibitors of IMPDH are also highly effective as immunosuppressive agents. The mechanism underlying these pleiotropic effects of depletion of guanine nucleotides is unknown. We have examined the effects of mycophenolic acid (MPA), a potent IMPDH inhibitor, on the cell cycle progression of activated normal human T lymphocytes. MPA treatment resulted in the inhibition of pRb phosphorylation and cell entry into S phase. The expression of cyclin D3, a major component of the cyclin-dependent kinase (CDK) activity required for pRb phosphorylation, was completely abrogated by MPA treatment of T cells activated by interleukin-2 (IL-2) and leucoagglutinin (PHA-L), whereas the expression of cyclin D2, CDK6, and CDK4 was more mildly attenuated. The direct kinase activity of a complex immunoprecipitated with anti-CDK6 antibody was also inhibited. In addition, MPA prevented the IL-2-induced elimination of p27(Kip1), a CDK inhibitor, and resulted in the retention of high levels of p27(Kip1) in IL-2/PHA-L-treated T cells bound to CDK2. These results indicate that inhibition of the de novo synthesis of guanine nucleotides blocks the transition of normal peripheral blood T lymphocytes from G0 to S phase in early- to mid-G1 and that this cell cycle arrest results from inhibition of the induction of cyclin D/CDK6 kinase and the elimination of p27(Kip1) inhibitory activity.
Article
The discovery of isozymes (types I and II) of IMP dehydrogenase (IMPDH; EC 1.1.1.205), the rate-limiting enzyme of de novo GTP biosynthesis, has attracted attention as a possible novel approach to cancer diagnosis and selective tumor cell chemotherapy. To elucidate differences in expression and regulation of the two IMPDH isozymes, we examined the steady-state levels of these mRNAs in various types of leukemic cells from patients. Northern blot analysis revealed that type II IMPDH was more active transcriptionally (1.5- to 5.1-fold) in all the leukemic cells examined than in normal lymphocytes, whereas type I expression was similar. The increased expression of type II mRNA in leukemic cells was closely linked with the increase in total IMPDH activity (r = 0.92). When leukemic cells from a patient with chronic granulocytic leukemia in blast crisis were separated into blast-rich and mature leukocyte-rich fractions, the expression of type II mRNA correlated positively with the population of immature leukemic cells, whereas type I expression was unchanged. Treatment of leukemic blasts with 12-O-tetradecanoyl-phorbol-13-acetate for 5 days resulted in a 90% decrease in the expression of type II mRNA with macrophage-like differentiation, while the expression of type I mRNA was relatively stable. These observations suggest that expression of type II IMPDH is stringently linked with immature characteristics of leukemic cells; thus, it should be a selective target for antileukemic chemotherapy.
Article
Mycophenolic acid (MPA), an inhibitor of inosine monophosphate dehydrogenase, in nanomolar concentrations blocks proliferative responses of cultured human, mouse and rat T lymphocytes and B lymphocytes to mitogens or in mixed lymphocyte reactions. The inhibitory effect of MPA on lymphocyte proliferation is reversed by addition to culture media of deoxyguanosine or guanosine but not by addition of deoxyadenosine or adenosine. The findings suggest that the principal mechanism of action of low concentrations of MPA is depletion of deoxyguanosine triphosphate which is required for DNA synthesis. In immunosuppressive doses, MPA does not affect the formation of IL-1 by LPS-activated human peripheral blood monocytes. Unlike cyclosporin A and FK-506, MPA does not inhibit the formation of IL-2 and the expression of the IL-2 receptor in mitogen-activated human T lymphocytes. MPA suppresses mixed lymphocyte reactions when added 3 days after their initiation. These findings suggest that MPA does not inhibit early responses of T and B lymphocytes to mitogenic or antigenic stimulation but blocks the cells at the time of DNA synthesis. The cytostatic effect of MPA is more potent on lymphocytes than on other cell types, such as fibroblasts and endothelial cells. MPA also inhibits antibody formation by polyclonally activated human B lymphocytes. MPA is an immunosuppressive agent reversibly inhibiting proliferation of T and B lymphocytes and antibody formation, with a profile of activity different from that of other immunosuppressive drugs. Human T and B lymphocytic and promonocytic cell lines are highly sensitive to the antiproliferative effects of MPA, whereas the erythroid precursor cell line K562 is less susceptible. The effect of MPA on cells of the monocyte-macrophage lineage could exert long-acting anti-inflammatory activity. MPA or analogues may have therapeutic utility in diseases such as rheumatoid arthritis, for prevention of allograft rejection and in lymphocytic or monocytic leukaemias and lymphomas.
Article
The immunosuppressive drug, mizoribine, has been used to prevent rejection of organ allografts in humans and in animal models. Based on studies in cell lines, mizoribine has been postulated to be an inhibitor of inosine monophosphate (IMP) dehydrogenase (EC1.2.1.14), a pivotal enzyme in the formation of guanine ribonucleotides from IMP. To further characterize the mechanism of action of this drug, we studied the effect of mizoribine on human peripheral blood T cells stimulated with alloantigen, anti-CD3 MAb, or pharmacologic mitogens. Mizoribine (1-50 micrograms/ml) was able to inhibit T cell proliferation by 10-100% in a dose-dependent fashion to all stimuli tested. Measurements of purine ribonucleotide pools by HPLC showed that mizoribine led to a decrease in intracellular GTP levels, and that repletion of GTP reversed its antiproliferative effects. We also examined sequential events occurring after T cell stimulation. Early events in T cell activation, as assessed by steady-state mRNA levels of c-myc, IL-2, c-myb, histone, and cdc2 kinase, as well as surface IL-2 receptor expression, were unaffected. However, cell cycle analysis revealed decreased numbers of cells in S, G2, and M phases, and showed that the G1/S block was reversed with GTP repletion. These data indicate that mizoribine has an effect on T cell proliferation by a mechanism distinct from that of cyclosporine or corticosteroids, and therefore may be useful in combination immunosuppressive regimens.
Article
Some monoclonal antibodies (mAbs) against interleukin (IL) 1 alpha have been found to activate antigen-presenting cells (APC, human peripheral blood monocytes and B lymphocytes), so that unstimulated T lymphocytes cultured with them are induced to proliferate and secrete IL-2. Control mAbs of the same isotypes and mAbs against IL-1 beta do not activate APC. In the absence of APC, mAbs against IL-1 alpha do not induce proliferation of T lymphocytes. Mitomycin C-treated activated APC still induce T-cell proliferation. Proliferation of T lymphocytes cannot be induced by culture supernatants and requires contact with APC activated by mAbs against IL-1 alpha. The observations imply that surface membrane IL-1 alpha can function as a triggering molecule on APC, which could play an important role in the initiation of immune responses by T lymphocytes.