ArticlePDF Available

Preferential apoptosis for primary human leukemic stem cells

Authors:

Abstract and Figures

Acute myelogenous leukemia (AML) is typically a disease of stem progenitor cell origin. Interestingly, the leukemic stem cell (LSC) shares many characteristics with normal hematopoietic stem cells (HSCs) including the ability to self-renew and a predominantly G(0) cell-cycle status. Thus, although conventional chemotherapy regimens often ablate actively cycling leukemic blast cells, the primitive LSC population is likely to be drug-resistant. Moreover, given the quiescent nature of LSCs, current drugs may not effectively distinguish between malignant stem cells and normal HSCs. Nonetheless, based on recent studies of LSC molecular biology, we hypothesized that certain unique properties of leukemic cells could be exploited to induce apoptosis in the LSC population while sparing normal stem cells. In this report we describe a strategy using treatment of primary AML cells with the proteasome inhibitor carbobenzoxyl-l-leucyl-l-leucyl-l-leucinal (MG-132) and the anthracycline idarubicin. Comparison of normal and leukemic specimens using in vitro culture and in vivo xenotransplantation assays shows that the combination of these two agents induces rapid and extensive apoptosis of the LSC population while leaving normal HSCs viable. Molecular genetic studies using a dominant-negative allele of inhibitor of nuclear factor kappaB (IkappaBalpha) demonstrate that inhibition of nuclear factor kappaB (NF-kappaB) contributes to apoptosis induction. In addition, gene-expression analyses suggest that activation of p53-regulated genes are also involved in LSC apoptosis. Collectively, these findings demonstrate that malignant stem cells can be preferentially targeted for ablation. Further, the data begin to elucidate the molecular mechanisms that underlie LSC-specific apoptosis and suggest new directions for AML therapy.
Content may be subject to copyright.
Preferential induction of apoptosis for primary human
leukemic stem cells
Monica L. Guzman, Carol F. Swiderski, Dianna S. Howard, Barry A. Grimes, Randall M. Rossi, Stephen J. Szilvassy,
and Craig T. Jordan*
Blood and Marrow Transplant Program, Markey Cancer Center, Division of HematologyOncology, University of Kentucky Medical Center,
Lexington, KY 40536
Edited by Mark T. Groudine, Fred Hutchinson Cancer Research Center, Seattle, WA, and approved October 22, 2002 (received for review August 2, 2002)
Acute myelogenous leukemia (AML) is typically a disease of stem
progenitor cell origin. Interestingly, the leukemic stem cell (LSC)
shares many characteristics with normal hematopoietic stem cells
(HSCs) including the ability to self-renew and a predominantly G
0
cell-cycle status. Thus, although conventional chemotherapy reg-
imens often ablate actively cycling leukemic blast cells, the prim-
itive LSC population is likely to be drug-resistant. Moreover, given
the quiescent nature of LSCs, current drugs may not effectively
distinguish between malignant stem cells and normal HSCs. None-
theless, based on recent studies of LSC molecular biology, we
hypothesized that certain unique properties of leukemic cells could
be exploited to induce apoptosis in the LSC population while
sparing normal stem cells. In this report we describe a strategy
using treatment of primary AML cells with the proteasome inhib-
itor carbobenzoxyl-
L-leucyl-L-leucyl-L-leucinal (MG-132) and the an-
thracycline idarubicin. Comparison of normal and leukemic speci-
mens using in vitro culture and in vivo xenotransplantation assays
shows that the combination of these two agents induces rapid
and extensive apoptosis of the LSC population while leaving
normal HSCs viable. Molecular genetic studies using a dominant-
negative allele of inhibitor of nuclear factor
B(I
B
) demonstrate
that inhibition of nuclear factor
B (NF-
B) contributes to apoptosis
induction. In addition, gene-expression analyses suggest that ac-
tivation of p53-regulated genes are also involved in LSC apoptosis.
Collectively, these findings demonstrate that malignant stem cells
can be preferentially targeted for ablation. Further, the data begin
to elucidate the molecular mechanisms that underlie LSC-specific
apoptosis and suggest new directions for AML therapy.
A
cute myelogenous leukemia (AML) is a serious and often
lethal form of hematologic cancer. Although the develop-
ment of better chemotherapy regimens has improved remission
induction and overall survival, relapse remains a common prob-
lem, especially among older patients andor patients with poor
prognosis cytogenetics (1). In recent years the clinical charac-
teristics of AML have become better understood in light of
studies elucidating the biological origins of the disease. Several
lines of evidence clearly indicate that AML is a disease of stem
or progenitor cell origin, and that the leukemic stem cell (LSC)
stands apart from more mature leukemic cells with its own set
of unique biological properties (2–9). Thus, although chemo-
therapeutic agents effectively ablate leukemic blast cells in a
majority of patients, the efficacy of LSC targeting is not known.
Indeed, it is attractive to speculate that failure to sustain durable
remission may be due to a drug-refractoryresistant malignant
stem cell population. Given the potentially critical role of stem
cells in both the genesis and perpetuation of AML, recent studies
have attempted to better characterize LSC properties (10–12).
Notably, although the immunophenotype of LSCs is similar to
normal hematopoietic stem cells (HSCs; CD34
, CD38
, HLA-
DR
), there are at least three antigens with expression that is
known to vary in malignant cells: CD90, CD117, and CD123
(7–9). Thus, it has been possible to prospectively identify and
isolate enriched LSC populations, which has allowed subsequent
studies to analyze the cell-cycle status and gene-expression
characteristics of normal vs. malignant stem cells (13–15). A
somewhat counterintuitive finding from cell-cycle studies is that
the LSC population seems mostly quiescent, which seems to be
true despite the often aggressive characteristics of leukemic
disease. Thus, the LSC should not be preferentially susceptible
to cycle-active chemotherapeutic agents.
Because standard chemotherapy approaches may not effec-
tively target the LSC population, we have attempted to charac-
terize molecular properties of the LSC that could be useful for
apoptosis induction. Data from our laboratory has shown re-
cently that nuclear factor
B (NF-
B) is constitutively activated
in the majority of primary AML specimens (15). Although
activation of NF-
B is a relatively common feature of many
cancers (16–18), a surprising aspect of our studies was the finding
that NF-
B is active also in quiescent LSC populations. Thus,
strategies to inhibit NF-
B, and thereby block growth and
survival pathways regulated by NF-
B, may represent a useful
approach to more durable AML therapy. With this concept in
mind, we examined several agents that inhibit NF-
B. One such
class of drugs that is being widely explored for cancer therapy is
proteasome inhibitors (19, 20). Proteasome inhibition blocks
degradation of the NF-
B regulator I
B
and results in loss of
NF-
B activity. Initial studies of primary AML cells have
demonstrated that treatment with the proteasome inhibitor
carbobenzoxyl-
L-leucyl-L-leucyl-L-leucinal (MG-132) causes
rapid inhibition of NF-
B and strongly induces apoptosis (15).
Thus, proteasome inhibition seems to be a promising strategy for
ablation of leukemic cells.
Another well documented mechanism of apoptosis induction
is mediated by activation of specific p53-regulated genes such as
Bax, GADD45, and p21
WAF/CIP
(21–23). This mechanism is
typically activated in response to DNA-damaging agents includ-
ing the anthracycline family of chemotherapy drugs (24, 25),
which are widely used for AML-induction therapy. Interestingly,
anthracyclines have also been shown to induce NF-
B activity
(26, 27), presumably as a cellular survival mechanism to resist
toxic effects of the drug. Therefore, we reasoned that anthra-
cycline treatment would increase cellular dependency on NF-
B
activity and thus further sensitize cells to the loss of NF-
B
activity induced by proteasome inhibitors. Consequently, in the
present study an analysis of proteasome inhibitors in combina-
tion with anthracyclines has been performed. The results of these
studies demonstrate that LSCs are extremely sensitive to a
combination of proteasome inhibition and anthracycline treat-
ment and undergo rapid and extensive apoptosis when briefly
cultured with these two agents. Importantly, normal HSCs show
This paper was submitted directly (Track II) to the PNAS office.
Abbreviations: AML, acute myelogenous leukemia; LSC, leukemic stem cell; HSC, hemato-
poietic stem cell; NF-
B, nuclear factor
B; I
B
, inhibitor of NF-
B; CB, umbilical cord blood;
MG-132, carbobenzoxyl-
L-leucyl-L-leucyl-L-leucinal; IDR, idarubicin; NOD, nonobese dia-
betic; SCID, severe combined immunodeficient; EMSA, electrophoretic mobility-shift assay;
I
B-SR, I
B superrepressor.
*To whom correspondence should be addressed at: Markey Cancer Center, 800 Rose Street,
Room CC412, Lexington, KY 40536-0093. E-mail: cjordan@uky.edu.
16220–16225
PNAS
December 10, 2002
vol. 99
no. 25 www.pnas.orgcgidoi10.1073pnas.252462599
little to no effect when treated with the same agents. Thus,
apoptosis can be induced in noncycling malignant stem cells
while sparing the normal HSC population.
Materials and Methods
Cell Isolation and Culture. AML cells, normal bone marrow, and
umbilical cord blood (CB) cells were obtained with informed
consent and processed as described (9, 15). In some cases,
marrow and CB cells were also obtained from the National
Disease Research Interchange (NDRI). Samples were subjected
to density-gradient separation to isolate mononuclear cells fol-
lowed by cryopreservation. As needed, samples were thawed and
used immediately. For in vitro studies, cells were cultured in
serum-free medium (28) for 1 h before the addition of MG-132
(Calbiochem) andor idarubicin (IDR, PharmaciaUpjohn).
When IDR and MG-132 where used in combination, cells were
incubated with IDR for 15 min prior the addition of MG-132.
Nonobese Diabetic (NOD)Severe Combined-Immunodeficient (SCID)
Mouse Assays.
NODSCID (NOD.CB17-Prdkdc ScidJ) mice
(The Jackson Laboratory) were exposed to 300 rad of
irradi-
ation from a
137
Cs source 1 day before transplantation. Cells to
be assayed were resuspended in 0.2 ml of PBS (GIBCO) with 2%
albumin and injected via the tail vein (510 million cells per
recipient). After 68 weeks, animals were killed, and bone
marrow was analyzed for the presence of human cells by using
flow cytometry.
Flow Cytometry. Cell-cycle and apoptosis analyses were per-
formed as described (15, 29). To analyze NODSCID mice,
marrow cells were blocked with the anti-Fc receptor antibody
2.4G2 and 25% human serum followed by labeling with human-
specific CD34-FITC and CD45-PE monoclonal antibodies
(PharMingen). Control samples consisted of marrow cells from
nontransplanted mice. To distinguish HLA-A2
and HLA-A2
populations, cells were labeled by using FITC-conjugated mono-
clonal MA2.1 (kindly provided by John Yannelli, Division of
HematologyOncology, University of Kentucky Medical
Center). Adenovirus-infected AML cells were labeled with
CD34-PE (PharMingen), and CD34
GFP
cells were isolated
by fluorescence-activated cell sorting (94% purity).
Adenovirus Vector Construction and Infection. Adenovirus vectors
used in this study were based on the AdBM5GFP vector from
Quantum Biotechnologies (Montreal). Details of the vector
construction will be described elsewhere. Briefly, the vector was
digested with AflII BglII to remove the major late promoter,
which then was replaced with a cytomegalovirus promoter. A
mutant allele of I
B
(serine to alanine at positions 32 and 36)
was then cloned immediately downstream of the cytomegalo-
virus promoter by using the BglII site. Virus was generated by
homologous recombination in 293 cells and purified by using
standard procedures (30). Infection of primary AML cells was
performed exactly as described by Howard et al. (31).
Electrophoretic Mobility-Shift Assay (EMSA), Immunoblot, and RNA
Analysis. EMSA analysis was performed as described (15). For
immunoblots, cells were harvested and analyzed as described by
Jordan et al. (9). Blots were probed with anti-p53 (clone DO-1)
from Santa Cruz Biotechnology and antiactin from Sigma (clone
AC-15). RNA was prepared by using Trizol (Invitrogen) per
manufacturer instructions. Five micrograms of RNA per sample
were analyzed by using the RiboQuant kit and the h-stress-1
multiprobe set (PharMingen) for RNase protection assays. Gels
were scanned by PhosphorImager (Molecular Dynamics), and
bands were quantitated by using Kodak
1D software.
Results
Proteasome Inhibition Preferentially Ablates Primitive AML Cells
in
Vitro
. To assess the effects of proteasome inhibition on primitive
quiescent cells, studies were performed initially by using multi-
parameter flow cytometry. In Fig. 1A, cell-cycle analysis shows
that primitive cells of both normal and leukemic origin (defined
by the immunophenotype CD34
CD38
CD123
and
CD34
CD38
CD123
, respectively) are predominantly in G
0
.
This is evident based on the lack of labeling with nuclear antigen
Ki-67, which is a common marker for entry into G
1
(32). Based
on our studies (15), tests were performed first by using the
proteasome inhibitor MG-132 at 1.0
M for 12 h. This treatment
readily induced apoptosis for AML cells, whereas normal
CD34
cells were almost entirely unaffected (as shown by
annexin-V labeling, Fig. 1B). Analysis of the 13 AML specimens
described in Table 1 showed an average viability of 26 18%
after 12 h of culture in MG-132. In contrast, parallel studies of
four normal CD34
cell specimens showed an average viability
of 89 5% when analyzed by using the same conditions. All
cultures were performed in serum-free medium and in the
absence of any exogenous cytokines.
Fig. 1. (A) Cell-cycle proles for normal and leukemic CD34
CD38
speci-
mens. BM, bone marrow. (B) Flow-cytometric prole of normal (Left) and
leukemic (Right) CD34
cells stained with annexin-V FITC and 7AAD after 12 h
of treatment with 1
M MG-132. Numbers indicate the percentage of viable
cells. DAPI, 4,6-diamidino-2-phenylindole.
Table 1. AML specimens
Case FAB type Cytogenetics
1 M4 46,XY,del(7)
2 M2 46,XY
3 M5 46,XX
4 M4 48,XY,8,13
5M1M2 46,XY
6 M4 46,XX
7 M5 45,X,Y
8 M5 46,XX
9 MDSAML 46,XY
10 M1 46,XX
11 M4 47,XY,9
12 M2 46,XY,t(9;19);t(8;21)
13 M4 46,XY,del(7)
Guzman et al. PNAS
December 10, 2002
vol. 99
no. 25
16221
MEDICAL SCIENCES
Because MG-132 demonstrated toxicity to phenotypically
primitive AML cells in vitro, we sought to further investigate
toxicity to the LSC compartment when proteasome inhibition
was combined with the commonly used chemotherapeutic an-
thracycline IDR. Studies have indicated that anthracyclines
induce NF-
B in malignant cells (27). Thus, we hypothesized that
anthracycline treatment might sensitize leukemic cells to the
effects of proteasome inhibition (i.e., loss of NF-
B activity).
Moreover, anthracyclines have been shown to increase protea-
some activity in leukemic cells, which also could increase sen-
sitivity to proteasome inhibition. To test this theory, both
MG-132 and the anthracycline IDR were first titrated to rela-
tively nontoxic doses of 0.25
M and 15 ngml, respectively (data
not shown). Each specimen listed in Table 1 then was cultured
for 12 h in low-dose MG-132 (0.25
M) andor IDR (15 ngml).
MG-132 treatment alone was only mildly toxic to the cells (mean
viability 73 9%). Similarly, treatment with IDR alone
also showed only a small effect on cell viability (mean viability
88 9%). However, the combination of both drugs synergized
strongly to induce robust apoptosis (mean viability 11 5%).
Fig. 2A shows a representative example of an AML specimen
(Upper) vs. a normal CB specimen (Lower) treated with the
low-dose MG-132IDR combination. As observed for MG-132
alone, the AML specimen was extremely sensitive to treatment
with MG-132IDR, whereas the normal specimen showed only
a slight loss in viability.
To examine effects on the most primitive cells more specifi-
cally, four AML and normal specimens were assayed for apo-
ptosis induction in the CD34
CD38
population. As shown in
Fig. 2B, the primitive AML compartment was also affected
dramatically by the combination of MG-132IDR, whereas the
normal CD34
cells were almost entirely resistant. These data
demonstrate that MG-132 in combination with IDR induces
apoptosis specifically in primitive AML cells, whereas normal
primitive cells seem largely unaffected.
Treatment with MG-132 and IDR Destroys LSCs Able to Engraft
NODSCID Mice. To determine whether stem cells defined by
in vivo functional assays were impaired by treatment with
MG-132IDR, normal and leukemic specimens were analyzed
by using transplantation into immune-deficient NODSCID
mice. NODSCID xenogeneic models have been used to assess
both human HSC and LSC activity (3335). This model can be
used therefore to identify therapies that affect either the LSC or
HSC population. Initially, cells from primary CB and AML
samples were treated with 0.25
M MG-132 and 15 ngml IDR
for 12 h and then injected into NODSCID mice. After 68
weeks, marrow was analyzed for the presence of donor cells by
using a human-specific antibody for CD45. Each triangle in Fig.
3A Left represents the percent engraftment for one animal
transplanted with untreated or MG-132IDR-treated CB cells
(15 animals in each group using CB from six independent
specimens). The data show that drug treatment does not impair
the capacity of normal HSCs to proliferate in NODSCID mice.
Further, analysis of human cells in animals receiving treated vs.
untreated CB cells showed no difference with respect to levels of
CD33 (myeloid), CD19 (B lymphoid), and CD34 (primitive)
cells, thereby indicating that the differentiation potential of
MG-132IDR-treated CB cells was not impaired (data not
Fig. 2. (A) Flow-cytometric analyses of primary AML cells (Upper) compared
with normal CB cells (Lower) after 18 h of treatment with or without 0.25
M
MG-132 15 ngml IDR. Numbers indicate the percentage of viable cells.
(B) Percentage of viable CD34
CD38
cells from primary AML (Left) and
normal CB (Right) cell samples after an 18-h treatment with 0.25
M MG-132
15 ngml IDR. Viability levels for each sample are normalized to untreated
control specimens.
Fig. 3. (A) Engraftment of NODSCID mice with CB cells after 12 or 18 h of
culture 0.25
M MG-132 and 15 ngml IDR (six independent CB specimens
were analyzed at each time point). (B) Engraftment of AML cells after 12 or
18 h of culture MG-132 and IDR (four independent AML samples were
analyzed after 12 h, and the two showing the highest levels of engraftment
were reanalyzed after 18 h of culture). Each triangle or circle represents a
single animal analyzed 6 8 weeks posttransplant for the percentage of
human cells in marrow. The median level of engraftment is indicated by the
horizontal bars.
16222
www.pnas.orgcgidoi10.1073pnas.252462599 Guzman et al.
shown). In some cases, CB samples lost their ability to engraft
after 12 h of culture without drugs. Notably, when the same CB
samples were treated with the MG-132IDR combination, they
often retained engraftment ability. Hence, the median engraft-
ment efficiency actually increased for drug-treated specimens
(median indicated by horizontal bars). A possible explanation for
this surprising result might be that the proteasome inhibitor
prevents degradation of proteins involved in homing. Alterna-
tively, the drug treatment may enhance survival or inhibit
terminal differentiation of normal stemprogenitor cells. Irre-
spective of the reason for this observation, the data indicate that
engraftment of normal cells is not impaired by MG-132IDR
treatment and supports the in vitro data suggesting the drugs are
not toxic to normal stem cells. In contrast, when AML samples
were treated in the same fashion and transplanted into NOD
SCID animals, engraftment efficiency was reduced dramatically.
Fig. 3B Left shows percent donor engraftment after 12 h of
culture drug treatment (25 animals in each group using AML
cells from four independent specimens). Two AML specimens
showed no detectable engraftment, and two specimens displayed
only low-level engraftment (3.97.5%) after they were exposed
to MG-132IDR. In an attempt to eradicate residual AML cells
more completely, we extended the drug exposure to 18 h for the
two specimens that showed engraftment at 12 h (Fig. 3B Right).
Longer exposure resulted in a further decrease in engraftment
but did not eliminate AML cells completely (0.12.8% engraft-
ment, seven animals in each group). Finally, to determine
whether the few remaining AML cells retained self-renewal
potential, marrow from primary animals was transplanted into
secondary NODSCID recipients (in the absence of any further
drug treatment). At 6 weeks posttransplant, no detectable
human cells were found in the marrow of secondary recipients,
thereby suggesting that the original drug treatment had ablated
all NODSCID engrafting activity completely. Control experi-
ments using normal CB samples were also performed by using
the 18-h MG-132IDR regimen (Fig. 3A Right). Although
overall engraftment of the CB samples was affected by 18 h of
culture, treatment with MG-132IDR did not reduce engraft-
ment efficiency further when compared with untreated controls
(15 animals per group).
Because treatment with MG-132IDR strongly induced apopto-
sis in the total AML cell population, it is possible that LSCs that
survive drug treatment might fail to engraft because of the presence
of excess dead cells. To address this possibility, an experiment was
performed in which CB and AML cells were treated with the
combination of MG-132IDR and coinjected into NODSCID
animals (n 6). If apoptotic cells induce an inhibitory effect on the
engraftment of viable stem cells, one would predict reduced en-
graftment when the MG-132IDR-treated cells are injected to-
gether. The specimens used for this experiment expressed disparate
HLA-A2 loci such that normal vs. AML cells could be distinguished
by flow cytometry. After 68 weeks, the animals were killed, and
the bone marrow was analyzed for engraftment of human cells. Fig.
4 shows a representative example of bone marrow cells stained with
human-specific CD45 and HLA-A2 antibodies. The total percent-
age of CD45
cells in both treated and untreated samples was very
similar (7379%). However, labeling with the HLA-A2 antibody
shows strong AML cell engraftment in the untreated controls (74%
of the CD45
population) but virtually no leukemic engraftment in
MG-132IDR-treated samples (0.5% of the CD45
population).
These data indicate that the inability of treated AML samples to
engraft in the animals is not due to an inhibitory effect caused by
apoptotic cells present in the transplanted sample. Taken together,
the data in Figs. 24 strongly suggest that the combination of
MG-132 and IDR effectively eradicates human LSCs while sparing
the normal HSC population.
Inhibition of NF-
B Contributes to Apoptosis Induced by MG-132IDR
Treatment.
To investigate the mechanisms that underlie LSC-
specific apoptosis, a series of molecular studies were performed.
Previously we showed that NF-
B is constitutively activated in
primary LSC populations but is not detected in normal CD34
cells (15). Further, these studies showed that treatment with 1.0
M MG-132 was sufficient to inhibit NF-
B activity. Thus,
EMSA analysis was used in the present studies to determine the
activity of NF-
B in AML cells treated with 0.25
M MG-132
and 15 ngml IDR. To assess molecular changes occurring at
early times posttreatment (i.e., before the onset of overt apo-
ptosis), cells were isolated after6hofdrugexposure. As shown
in Fig. 5A, treatment with MG-132 IDR induced strong
inhibition of NF-
B in comparison to untreated controls in five
independent AML specimens. This observation suggests that
NF-
B might mediate survival in primary AML cells. To address
this possibility directly, an adenovirus vector was constructed
that encodes both a mutant version of I
B
and the GFP gene.
The I
B
gene used for this vector encodes an allele known as
the I
B superrepressor (I
B-SR). This gene is mutated at serines
32 and 36 such that it cannot be phosphorylated and hence is not
degraded by normal proteasomal mechanisms. Thus, I
B-SR
expression exerts potent inhibition of NF-
B activity as a result
of stabilized I
B
protein levels. Three primary AML specimens
were transduced with the Ad-I
B-SR vector (Ad-I
B) or a
control virus encoding GFP alone (Ad-GFP) as described (31).
Infected populations then were monitored over time to assess
viability of CD34
blast cells. The data in Fig. 5C show that
although Ad-GFP-infected cells maintained 100% viability over
a 36-h period relative to uninfected controls, cells infected with
Ad-I
B-SR decreased to 50% of control values (52 4%
viable cells). EMSA analysis demonstrated that expression of
I
B-SR by the vector completely inhibited NF-
B activity in
transduced AML cells (Fig. 5D). These data indicate that NF-
B
activity is important for survival of primary AML cells, but that
inhibition of NF-
B alone is not sufficient to mediate the very
rapid and extensive apoptosis observed after treatment with
MG-132IDR.
Activation of a p53-Regulated Mechanism During Apoptosis Induced
by MG-132IDR.
To further characterize molecular mechanisms
related to apoptosis induction, pathways commonly associated
with cell death were examined. Recent studies by Tergaonkar et
al. (36) have shown that treatment with the anthracycline
doxorubicin induces a p53-mediated apoptosis response. More-
over, this report showed that loss of NF-
B activity resulted in
Fig. 4. AML and CB cells were cultured for 12 h 0.25
M MG-132 and 15
ngml IDR and injected into sublethally irradiated NODSCID mice. Bone
marrow was analyzed 6 weeks later with anti-human CD45-PE and HLA-A2-
FITC to detect engraftment of human cells. The histograms indicate the
percent of human AML vs. CB cells.
Guzman et al. PNAS
December 10, 2002
vol. 99
no. 25
16223
MEDICAL SCIENCES
a stronger induction of apoptosis after activation of p53. Thus,
we reasoned that the IDR used in our studies might elicit a
similar response. To test this hypothesis, total protein and RNA
were prepared from cells cultured for6hintheabsence or
presence of 0.25
M MG-132 and 15 ngml IDR. Immunoblots
(Fig. 5B) showed that levels of p53 protein were increased
strongly by drug treatment in six independent AML specimens.
This observation suggests that p53-mediated changes in gene
expression might be involved in apoptosis induced by MG-132
IDR treatment. To examine this question in more detail, RNA
was isolated from five primary AML specimens after6hof
culture and expression of the p53-regulated genes Bax, GADD45,
and p21
WAF/CIP
were analyzed by RNase protection assays. These
three genes are strongly associated with p53-induced apoptosis
in a variety of different cell types (2123). As shown in Fig. 5E,
RNA levels of Bax, GADD45, and p21
WAF/CIP
all are increased
by treatment with MG-132IDR. Interestingly, the antiapoptotic
protein Bcl-xL was reduced markedly in cells treated with
MG-132 IDR. Bcl-xL is an NF-
B-regulated gene; thus the
observed decrease in Bcl-xL mRNA is consistent with the block
to NF-
B activity seen in Fig. 5A. Collectively, these data suggest
that the combination of MG-132 and IDR induces apoptosis
through activation of p53-regulated genes and simultaneous
inhibition of NF-
B.
Discussion
Recent studies clearly indicate a central role for LSCs in the
pathogenesis of AML and emphasize the need to develop
treatment strategies that specifically target this rare population
(12). Equally important, it is necessary to identify approaches
that minimize toxicity to the normal HSC, which is required to
sustain all blood-cell lineages. Consequently, we sought to
investigate drug-treatment regimens that would target the LSC
population specifically and preferentially. To this end, we have
demonstrated that low concentrations of MG-132 (0.25
M) in
combination with IDR (15 ngml) are sufficient to induce a
strong apoptotic response in primary AML specimens during a
short in vitro culture period (12 h). More importantly, by using
flow-cytometric analysis of phenotypically primitive cells and
functional assays in NODSCID mice, the data indicate that
MG-132 IDR also effectively ablates the LSC population. In
contrast, normal HSCs are almost entirely refractory to the same
treatment. These findings formally demonstrate that with the
appropriate stimulus quiescent LSCs are preferentially suscep-
tible to apoptosis induction. Thus, it should be possible to
develop treatment regimens that specifically target the LSC
population. Moreover, recent clinical evaluation of pharmaceu-
tical-grade proteasome inhibitors (37) may soon provide an
opportunity to examine the therapeutic potential of such drugs
in combination with anthracyclines.
Going forward, it will be important to develop a detailed
profile of the molecular events that mediate LSC-specific apo-
ptosis. Studies in this report investigate this phenomenon by
analyzing unique molecular characteristics of the LSC popula-
tion. Previously it was shown that primitive AML cells exhibit
substantial NF-
B activity (15). Given the known role of NF-
B
in the growth and survival of many tumor types (16), we
speculated that inhibition of this factor might be a key step
toward inducing LSC apoptosis. Interestingly, although our
EMSA data show that NF-
B is inhibited potently by MG-132
IDR treatment, subsequent studies using a dominant-negative
allele of I
B
showed that down-regulation of NF-
B activity
alone was not sufficient to mediate the same degree of rapid and
extensive apoptosis in AML cells as was observed for MG-132
IDR. This finding indicates that other effects of the drug
treatment must be contributing to AML cell death. In seeking to
characterize LSC apoptosis further, we hypothesized that pro-
teasome inhibition might be stabilizing proteins relevant to
apoptosis induction such as p53 (3840). Moreover, the activity
of IDR was expected to activate p53-mediated apoptosis mech-
anisms. Immunoblot studies confirmed that p53 was increased in
drug-treated cells. Further investigation by RNase protection
assay showed that several p53-regulated genes known to be
mediators of apoptosis were up-regulated in the MG-132IDR-
treated cells. These data indicate that activation of a p53-
dependent mechanism is likely to be one component of the
overall apoptosis process. Notably, p53 is only mutated in 9%
of AML specimens (41); thus a strategy that relies on functional
p53 for ablation of leukemic cells should be applicable to a
majority of patients. Future studies may directly address the role
of p53 by expressing a dominant-negative allele of the gene in
Fig. 5. (A) NF-
B EMSA of nuclear extracts from ve primary AML specimens
after6hofculture (UNT., untreated; MGIDR, 0.25
M MG-132 15 ngml
IDR). Numbers above each lane indicate the specimen number. (B) Immuno-
blot analysis of p53 for each of the specimens shown in A. Each blot was
stripped and reprobed with actin (lower panel of each p53 blot). (C) Three
primary AML specimens were infected with Ad-GFP or Ad-I
B-SR and cultured
for 36 h. The graph indicates the average percentage of viable GFP
cells
where Ad-GFP is dened as 100%. (D) EMSA analysis of NF-
B in sorted GFP
or GFP
cells at 12 h postinfection with Ad-GFP or Ad-I
B-SR (lane 1, NF-
B
consensus probe; lane 2, probe 100-fold excess unlabeled consensus probe).
(E) Relative change in expression level as determined by RNase protection
assay for ve primary AML specimens after6hofculture in 0.25
M MG-132
15 ngml IDR. Untreated specimens were assigned an arbitrary level of 1.0
(shown by the horizontal bar). Analysis of each specimen was performed in
triplicate (standard deviation is shown by error bars). Loading was normalized
by using L32 ribosomal and glyceraldehyde-3-phosphate dehydrogenase
probes as controls.
16224
www.pnas.orgcgidoi10.1073pnas.252462599 Guzman et al.
AML cells and determining whether loss of p53 activity is
sufficient to block apoptosis induced by MG-132IDR.
Another important issue in understanding the biology of LSCs
is determining how NF-
B is activated. Our studies indicate that
NF-
B plays an important role in the survival of LSCs and
represents a potentially useful target for therapeutic interven-
tion. One mechanism for NF-
B activation may be related to
mutation of the Flt3 gene. Constitutive activation of Flt3 has
emerged recently as the most commonly known aberration in
AML (42). Studies have shown that signaling via Flt3 can
stimulate Ras (43), which in turn is a known activator of NF-
B
(16). Thus, pathways leading from Flt3 to NF-
B seem to exist.
Moreover, several recent studies have begun to examine the use
of Flt3 inhibitors for AML therapy (44). Consequently, direct
analysis of the degree to which Flt3- and NF-
B-regulated
pathways overlap are now feasible. Interestingly, the kinetics and
degree of primary AML cell death reported for Flt3 inhibitors
(45, 46) appear very similar to data in this report using expres-
sion of I
B-SR (i.e., 4050% cell death over 3672hof
treatment). This similarity may indicate a substantial degree of
overlap between strategies that target Flt3 and NF-
B and
suggests that Flt3 inhibitors and proteasome inhibitors may be
functionally equivalent in the context of leukemic cell biology. If
this concept is true, then based on findings in this study, we
suggest that combining Flt3 inhibition with IDR treatment may
yield a more rapid and robust induction of apoptosis, as was
observed for MG-132 IDR. This approach may be attractive
for clinical use, because Flt3 inhibitors are expected to have less
nonspecific toxicity than proteasome inhibitors. Further, we
suggest that combination of Flt3 inhibitors with agents such as
IDR may increase the likelihood that they will target quiescent
LSC populations effectively.
We thank Drs. Katherine Borden, Gary Van Zant, Hartmut Geiger, and
Deborah Echlin for critical evaluation of the manuscript. We also thank
Dr. Marty Mayo for helpful discussions and for providing the I
B-SR
gene. We gratefully acknowledge the generous support of The Markey
Cancer Center Foundation and The Donatina Colachicco Cancer Re-
search Fund. This work was supported by American Cancer Society
Grant RPG-99-206-01-LBC (to C.T.J.) and National Institutes of Health
Grant R01-CA90446 (to C.T.J.). D.S.H. is a fellow of the Abraham J. and
Phyllis Katz Foundation.
1. Lowenberg, B., Downing, J. R. & Burnett, A. (1999) N. Engl. J. Med. 341,
10511062.
2. Fialkow, P. J., Singer, J. W., Adamson, J. W., Vaidya, K., Dow, L. W., Ochs,
J. & Moohr, J. W. (1981) Blood 57, 10681073.
3. Fialkow, P. J., Singer, J. W., Raskind, W. H., Adamson, J. W., Jacobson, R. J.,
Bernstein, I. D., Dow, L. W., Najfeld, V. & Veith, R. (1987) N. Engl. J. Med.
317, 468473.
4. Lapidot, T., Sirard, C., Vormoor, J., Murdoch, B., Hoang, T., Caceres-Cortes,
J., Minden, M., Paterson, B., Caligiuri, M. A. & Dick, J. E. (1994) Nature 367,
645648.
5. Bonnet, D. & Dick, J. E. (1997) Nat. Med. 3, 730737.
6. Blair, A., Hogge, D. E. & Sutherland, H. J. (1998) Blood 92, 4325 4335.
7. Blair, A., Hogge, D. E., Ailles, L. E., Lansdorp, P. M. & Sutherland, H. J. (1997)
Blood 89, 31043112.
8. Blair, A. & Sutherland, H. J. (2000) Exp. Hematol. (Charlottesville, Va) 28,
660671.
9. Jordan, C. T., Upchurch, D., Szilvassy, S. J., Guzman, M. L., Howard, D. S.,
Pettigrew, A. L., Meyerrose, T., Rossi, R., Grimes, B., Rizzieri, D. A., et al.
(2000) Leukemia 14, 17771784.
10. Sutherland, H. J., Blair, A. & Zapf, R. W. (1996) Blood 87, 4754 4761.
11. Ailles, L. E., Gerhard, B. & Hogge, D. E. (1997) Blood 90, 25552564.
12. Jordan, C. T. (2002) Leukemia 16, 559562.
13. Terpstra, W., Ploemacher, R. E., Prins, A., van Lom, K., Pouwels, K., Wognum,
A. W., Wagemaker, G., Lowenberg, B. & Wielenga, J. J. (1996) Blood 88,
19441950.
14. Guzman, M. L., Upchurch, D., Grimes, B., Howard, D. S., Rizzieri, D. A.,
Luger, S. M., Phillips, G. L. & Jordan, C. T. (2001) Blood 97, 21772179.
15. Guzman, M. L., Neering, S. J., Upchurch, D., Grimes, B., Howard, D. S.,
Rizzieri, D. A., Luger, S. M. & Jordan, C. T. (2001) Blood 98, 23012307.
16. Mayo, M. W. & Baldwin, A. S. (2000) Biochim. Biophys. Acta 1470, M55M62.
17. Garg, A. & Aggarwal, B. B. (2002) Leukemia 16, 10531068.
18. Karin, M., Cao, Y., Greten, F. R. & Li, Z. W. (2002) Nat. Rev. Cancer 2,
301310.
19. Hallahan, D. E. & Teng, M. (2000) Int. J. Radiat. Oncol. Biol. Phys. 47, 859860.
20. Almond, J. B. & Cohen, G. M. (2002) Leukemia 16, 433443.
21. Miyashita, T., Krajewski, S., Krajewska, M., Wang, H. G., Lin, H. K.,
Liebermann, D. A., Hoffman, B. & Reed, J. C. (1994) Oncogene 9, 17991805.
22. Kastan, M. B., Zhan, Q., el-Deiry, W. S., Carrier, F., Jacks, T., Walsh, W. V.,
Plunkett, B. S., Vogelstein, B. & Fornace, A. J., Jr. (1992) Cell 71, 587597.
23. el-Deiry, W. S., Tokino, T., Velculescu, V. E., Levy, D. B., Parsons, R., Trent,
J. M., Lin, D., Mercer, W. E., Kinzler, K. W. & Vogelstein, B. (1993) Cell 75,
817825.
24. Lowe, S. W., Bodis, S., McClatchey, A., Remington, L., Ruley, H. E., Fisher,
D. E., Housman, D. E. & Jacks, T. (1994) Science 266, 807810.
25. Lowe, S. W., Ruley, H. E., Jacks, T. & Housman, D. E. (1993) Cell 74, 957967.
26. Laurent, G. & Jaffrezou, J. P. (2001) Blood 98, 913924.
27. Boland, M. P., Foster, S. J. & ONeill, L. A. (1997) J. Biol. Chem. 272,
1295212960.
28. Lansdorp, P. M. & Dragowska, W. (1992) J. Exp. Med. 175, 15011509.
29. Jordan, C. T., Yamasaki, G. & Minamoto, D. (1996) Exp. Hematol. (Char-
lottesville, Va) 24, 13471355.
30. Prevec, L. & Graham, F. L. (1991) Methods in Molecular Biology: Gene Transfer
and Expression Protocols (Humana, Clifton, NJ).
31. Howard, D. S., Rizzierri, D., Grimes, B., Upchurch, D., Phillips, G., Stewart,
A., Yannelli, J. & Jordan, C. T. (1999) Leukemia 13, 16081616.
32. Gerdes, J., Lemke, H., Baisch, H., Wacker, H. H., Schwab, U. & Stein, H.
(1984) J. Immunol. 133, 17101715.
33. Dick, J. E. (1996) Semin. Immunol. 8, 197206.
34. Dick, J. E., Bhatia, M., Gan, O., Kapp, U. & Wang, J. C. (1997) Stem Cells
(Dayton) 15, 199203.
35. Lapidot, T., Fajerman, Y. & Kollet, O. (1997) J. Mol. Med. 75, 664673.
36. Tergaonkar, V., Pando, M., Vafa, O., Wahl, G. & Verma, I. (2002) Cancer Cell
1, 493503.
37. Adams, J. (2002) Curr. Opin. Chem. Biol. 6, 493500.
38. Dietrich, C., Bartsch, T., Schanz, F., Oesch, F. & Wieser, R. J. (1996) Proc. Natl.
Acad. Sci. USA 93, 1081510819.
39. Lopes, U. G., Erhardt, P., Yao, R. & Cooper, G. M. (1997) J. Biol. Chem. 272,
1289312896.
40. Naujokat, C., Sezer, O., Zinke, H., Leclere, A., Hauptmann, S. & Possinger, K.
(2000) Eur. J. Haematol. 65, 221236.
41. Stirewalt, D. L., Kopecky, K. J., Meshinchi, S., Appelbaum, F. R., Slovak, M. L.,
Willman, C. L. & Radich, J. P. (2001) Blood 97, 35893595.
42. Gilliland, D. G. & Griffin, J. D. (2002) Curr. Opin. Hematol. 9, 274281.
43. Mizuki, M., Fenski, R., Halfter, H., Matsumura, I., Schmidt, R., Muller, C.,
Gruning, W., Kratz-Albers, K., Serve, S., Steur, C., et al. (2000) Blood 96,
39073914.
44. Gilliland, D. G. & Griffin, J. D. (2002) Blood 100, 15351542.
45. Levis, M., Tse, K. F., Smith, B. D., Garrett, E. & Small, D. (2001) Blood 98,
885887.
46. Levis, M., Allebach, J., Tse, K. F., Zheng, R., Baldwin, B. R., Smith, B. D.,
Jones-Bolin, S., Ruggeri, B., Dionne, C. & Small, D. (2002) Blood 99,
38853891.
Guzman et al. PNAS
December 10, 2002
vol. 99
no. 25
16225
MEDICAL SCIENCES
... HOIP, HOIL-1L, and SHARPIN are highly expressed in several cancers, including diffuse large B-cell lymphoma (DLBCL), and play active roles in tumor progression and drug resistance mainly through activation of the nuclear factor kappa B (NF-κB) pathway [12][13][14][15][16][17][18]. The NF-κB pathway has been reported to be constitutively active in several cancers including myeloid leukemia [19], particularly in leukemia stem cell (LSC) fractions [20][21][22]. Given that the NF-κB pathway is constitutively activated in LSC fractions, inhibition of LUBAC could be a promising strategy for treating myeloid leukemia. ...
... Deletion of Hoip impairs propagation of myeloid leukemia through inhibition of NF-κB pathway LUBAC generally activates the canonical NF-κB pathway [4][5][6][7], and the NF-κB pathway is constitutively active in several cancers including AML [19][20][21][22]. We, thus, evaluated the protein levels of Iκbα and of p65 phosphorylation on Ser536 (p-p65) in myeloid leukemia cells by flow cytometric analysis. ...
... Consistent with the role of LUBAC in the canonical NF-κB pathway in several cell types [4][5][6][7], we also found that deletion or knockdown of Hoip led to the inhibition of Iκbα degradation and a reduction in p-p65 in mouse and human myeloid leukemia cells. In fact, the role of the NF-κB pathway has been reported in various types of cancers, including myeloid leukemia [19][20][21][22]. Notably, the NF-κB pathway has been shown to be constitutively activated in murine and human LSC fractions in AML [20][21][22] and is maintained through autocrine TNF-α secretion [22], suggesting that inhibition of LUBAC may block the positive feedback of NF-κB/TNF-α signaling in LSCs. ...
Article
Full-text available
We investigated the role of Hoip, a catalytic subunit of linear ubiquitin chain assembly complex (LUBAC), in adult hematopoiesis and myeloid leukemia by using both conditional deletion of Hoip and small-molecule chemical inhibitors of Hoip. Conditional deletion of Hoip led to significantly longer survival and marked depletion of leukemia burden in murine myeloid leukemia models. Nevertheless, a competitive transplantation assay showed the reduction of donor-derived cells in the bone marrow of recipient mice was relatively mild after conditional deletion of Hoip. Although both Hoip-deficient hematopoietic stem cells (HSCs) and leukemia stem cells (LSCs) impaired the maintenance of quiescence, conditional deletion of Hoipinduced apoptosis in LSCs but not HSCs in vivo. Structure-function analysis revealed that LUBAC ligase activity and the interaction of LUBAC subunits were critical for the propagation of leukemia. Hoip regulated oxidative phosphorylation pathway independently of nuclear factor kappa B pathway in leukemia, but not in normal hematopoietic cells. Finally, the administration of thiolutin, which inhibits the catalytic activity of Hoip, improved the survival of recipients in murine myeloid leukemia and suppressed propagation in the patient-derived xenograft model of myeloid leukemia. Collectively, these data indicate that inhibition of LUBAC activity may be a valid therapeutic target for myeloid leukemia.
... GSEA analysis revealed that signaling pathways of NF-κB and KRAS were significantly activated in HRPSgroup. AS a member of transcription factors, NF-κB is widely involved in many biological processes, such as responses of immune and inflammation, proliferation, differentiation, cell survival, and all of which were strongly linked to survival of AML stem cells [27][28][29][30][31]. ...
Article
To explore effects of aging-related genes (ARGs) on the prognosis of Acute Myeloid Leukemia (AML), a seven-ARGs signature was developed and validated in AML patients. The numbers of seven-ARG sequences were selected to construct the survival prognostic signature in TCGA-LAML cohort, and two GEO datasets were used independently to verify the prognostic values of signature. According to seven-ARGs signature, patients were categorized into two subgroups. Patients with high-risk prognostic score were defined as HRPS-group/high-risk group, while others were set as LRPS-group/low-risk group. HRPS-group presented adverse overall survival (OS) than LRPS-group in TCGA-AML cohort (HR=3.39, P<0.001). In validation, the results emphasized a satisfactory discrimination in different time points, and confirmed the poor OS of HRPS-group both in GSE37642 (HR=1.96, P=0.001) and GSE106291 (HR=1.88, P<0.001). Many signal pathways, including immune- and tumor-related processes, especially NF-κB signaling, were highly enriched in HRPS-group. Coupled with high immune-inflamed infiltration, the HRPS-group was highly associated with the driver gene and oncogenic signaling pathway of TP53. Prediction of blockade therapy targeting immune checkpoint indicated varied benefits base on the different ARGs signature score, and the results of predicted drug response suggested that Pevonedistat, an inhibitor of NEDD8-activating enzyme, targeting NF-κB signaling, may have potential therapeutic value for HRPS-group. Compared with clinical factors alone, the signature had an independent value and more predictive power of AML prognosis. The 7-ARGs signature may help to guide clinical-decision making to predict drug response, and survival in AML patients.
... Cancer-initiating cells, often called cancer stem cells, are a subpopulation of cancer cells with stem cell-like properties that have recently been identified in a wide range of human cancers, including BC, prostate, brain, liver, pancreas, and blood cancer (159)(160)(161)(162)(163)(164)(165)(166)(167)(168)(169). These cells are resistant to several chemotherapy protocols and are considered the leading cause of cancer recurrence after treatment (170)(171)(172). However, some biological mechanisms, including overexpression of ATP-binding cassette transporters, and increasing anti-apoptotic and effective DNA damage response, are associated with chemoresistance in cancer cells; None of these processes are stem cell-like properties, and thus their contributions to tumor-initiating cell tolerance therapies remain unresolved (173). ...
Article
Full-text available
A growing body of evidence has revealed that microRNA (miRNA) expression is dysregulated in cancer, and they can act as either oncogenes or suppressors under certain conditions. Furthermore, some studies have discovered that miRNAs play a role in cancer cell drug resistance by targeting drug-resistance-related genes or influencing genes involved in cell proliferation, cell cycle, and apoptosis. In this regard, the abnormal expression of miRNA-128 (miR-128) has been found in various human malignancies, and its verified target genes are essential in cancer-related processes, including apoptosis, cell propagation, and differentiation. This review will discuss the functions and processes of miR-128 in multiple cancer types. Furthermore, the possible involvement of miR-128 in cancer drug resistance and tumor immunotherapeutic will be addressed.
Article
Full-text available
Several studies suggest Valproate's (VPA) therapeutic use in treating seizures, epilepsy, and bipolar disorder. Valproate is a class I histone deacetylases (HDACs) inhibitor and an attractive chemotherapeutic agent for targeting cancer. Few reports suggest that Valproate can suppress cell growth and cell differentiation and is linked with anti‐tumor activity. However, Valproate‐associated anti‐tumoral function and intracellular signaling cascade‐mediated anti‐cellular proliferation activities still need to be better understood. This current study suggests that Valproate can elevate proteasomal dysfunctions, resulting in misfolded protein accumulation and abnormal mitochondrial functions that successively induce apoptosis. Present findings indicate that treatment of Valproate inhibits Proteasome activities and also aggravates accumulation of expanded polyglutamine proteins and other proteasomal substrates. Overall, the aggregation of aberrant proteins and mitochondrial dysfunctions are observed, such as cytochrome c release, and disturbed mitochondrial membrane potential. Treatment of Valproate induces apoptotic morphological changes and cell death. These observations suggest that Valproate can cause mitochondrial abnormalities associated with apoptosis. These findings can provide new possible insights and suggest molecular approaches for developing better and specific Proteasome inhibitors that can be better useful with other anti‐tumor drugs for treatment of cancer and other complex diseases.
Article
Full-text available
The current scientific literature has extensively explored the potential role of proteasome inhibitors (PIs) in the NF-κB pathway of leukemia and lymphoma. The ubiquitin-proteasome system (UPS) is a critical component in regulating protein degradation in eukaryotic cells. PIs, such as BTZ, are used to target the 26S proteasome in hematologic malignancies, resulting in the prevention of the degradation of tumor suppressor proteins, the activation of intrinsic mitochondrial-dependent cell death, and the inhibition of the NF-κB signaling pathway. NF-κB is a transcription factor that plays a critical role in the regulation of apoptosis, cell proliferation, differentiation, inflammation, angiogenesis, and tumor migration. Despite the successful use of PIs in various hematologic malignancies, there are limitations such as resistant to these inhibitors. Some reports suggest that PIs can induce NF-κB activation, which increases the survival of malignant cells. This article discusses the various aspects of PIs’ effects on the NF-κB pathway and their limitations. Video Abstract Supplementary Information The online version contains supplementary material available at 10.1186/s12964-023-01433-5.
Article
Full-text available
The role of tumor interaction with stromal components during carcinogenesis is crucial for the design of efficient cancer treatment approaches. It is widely admitted that tumor hypoxic stress is associated with tumor aggressiveness and thus impacts susceptibility and resistance to different types of treatments. Notable biological processes that hypoxia functions in include its regulation of tumor heterogeneity and plasticity. While hypoxia has been reported as a major player in tumor survival and dissemination regulation, the significance of hypoxia inducible factors in cancer stem cell development remains poorly understood. Several reports indicate that the emergence of cancer stem cells in addition to their phenotype and function within a hypoxic tumor microenvironment impacts cancer progression. In this respect, evidence showed that cancer stem cells are key elements of intratumoral heterogeneity and more importantly are responsible for tumor relapse and escape to treatments. This paper briefly reviews our current knowledge of the interaction between tumor hypoxic stress and its role in stemness acquisition and maintenance. Our review extensively covers the influence of hypoxia on the formation and maintenance of cancer stem cells and discusses the potential of targeting hypoxia-induced alterations in the expression and function of the so far known stem cell markers in cancer therapy approaches. We believe that a better and integrated understanding of the effect of hypoxia on stemness during carcinogenesis might lead to new strategies for exploiting hypoxia-associated pathways and their targeting in the clinical setting in order to overcome resistance mechanisms. More importantly, at the present time, efforts are oriented towards the design of innovative therapeutical approaches that specifically target cancer stem cells.
Article
Modulating the immune system to treat diseases, including myeloid malignancies, has resulted in the development of a multitude of novel therapeutics in recent years. Myelodysplastic syndromes or neoplasms (MDS) and acute myeloid leukemia (AML) are hematologic malignancies that arise from defects in hematopoietic stem and progenitor cells (HSPCs). Dysregulated immune responses, especially in innate immune and inflammatory pathways, are highly associated with the acquisition of HSPC defects in MDS and AML pathogenesis. In addition to utilizing the immune system in immunotherapeutic interventions such as CAR-T cell therapy, vaccines, and immune checkpoint inhibitors, mitigating dysregulation of innate immune and inflammatory responses in MDS and AML remains a priority in slowing the initiation and progression of these myeloid malignancies. This review provides a comprehensive summary of the current progress of diverse strategies to modulate the immune system in the treatment of MDS and AML.
Article
Full-text available
Introduction. The properties of progenitor cells in the stromal microenvironment, i.e. multipotent mesenchymal stromal cells (MMSC) and fi broblast colony-forming units (CFU-F), undergo changes in patients with chronic myelogenous leukaemia (CML). Aim. To compare the progenitor cells of the stromal microenvironment (MMSCs and CFU-Fs) obtained from the bone marrow of CML patients at the onset of the disease, one year after the start of the treatment and during the long-term treatment with tyrosine kinase inhibitors (TKI). Materials and methods. The study involved an analysis of the characteristics of MMSCs, the concentration of CFU-Fs in the bone marrow of CML patients, as well as the relative expression level of genes (REL) associated with differentiation and involved in the regulation of haematopoiesis. The analysis was performed at the onset of the disease, one year after the start of the treatment, as well as 3–8 and 9–16 years after the TKI therapy. MMSCs and CFU-Fs of healthy donors were used for control purposes. Results. The concentration of CFU-Fs at the onset of the disease did not differ from that in donors; however, the relative expression level of genes associated with differentiation was increased in the CFU-F colonies. A year after the start of TKI treatment, the concentration of CFU-Fs decreased by four times. Subsequently, the concentration increased to reach normal values following 8 years of TKI treatment. The total production of MMSCs was not changed at the onset of the disease; however, it decreased after a year of TKI treatment, subsequently returning to normal. The expression of many genes was altered in the MMSCs of patients, i.e. the REL of LIF and JAG1 increased by 10 and 2 times, respectively; in the course of treatment, the REL of LIF in MMSCs decreased, always remaining higher than in those of the donors, whereas the expression of JAG1 returned to normal. At the onset of the disease, the REL of LIF in the MMSCs of patients, who achieved a deep molecular response (DMR) within 17 months of the treatment, was three times lower than in the MMSCs of those patients who did not reach DMR within 50 months, with JAG1 not differing from that of donors. Conclusion. Changes in stromal progenitor cells are associated with the influence of tumour cells, as well as with TKI therapy. A normal expression level of JAG1 and a decreased expression level of LIF in the MMSCs of CML patients at the onset of the disease may be predictive of DMR achievement. Conflict of interest : the authors declare no conflict of interest. Financial disclosure : the study had no sponsorship.
Article
Full-text available
Recent studies suggest that the population of malignant cells found in human acute myelogenous leukemia (AML) arises from a rare population of leukemic stem cells (LSCs). LSCs have been documented for nearly all AML subtypes and have been phenotypically described as CD34+/CD38- or CD34+/HLA-DR-. Given the potentially critical role of these primitive cells in perpetuating leukemic disease, we sought to further investigate their molecular and cellular characteristics. Flow cytometric studies using primary AML tissue showed that the interleukin-3 receptor alpha chain (IL-3Ralpha or CD123) was strongly expressed in CD34+/CD38- cells (98 +/- 2% positive) from 16 of 18 primary specimens. Conversely, normal bone marrow derived CD34+/CD38- cells showed virtually no detectable expression of the CD123 antigen. To assess the functional role of IL-3Ralpha positive cells, purified CD34+/CD123+ leukemia cells were transplanted into immune deficient NOD/SCID mice. These experiments showed that CD123+ cells were competent to establish and maintain leukemic populations in vivo. To begin to elucidate a biological role for CD123 in leukemia, primary AML samples were analyzed with respect to signal transduction activity in the MAPK, Akt, and Stat5 pathways. Phosphorylation was not detected in response to IL-3 stimulation, thereby suggesting CD123 is not active in conventional IL-3-mediated signaling. Collectively, these data indicate that CD123 represents a unique marker for primitive leukemic stem cells. Given the strong expression of this receptor on LSCs, we propose that targeting of CD123 may be a promising strategy for the preferential ablation of AML cells.
Article
Full-text available
Adenoviruses have been isolated from a large number of different species (mammalian and fowl) and over 100 different serotypes have been reported, some 43 of them human. The human adenoviruses, particularly types 2, 5, and 12, have been the most extensively characterized, and these viruses have served as valuable tools in the study of the molecular biology of DNA replication, transcription, RNA processing, and protein synthesis in mammalian cells. Seeref. 1 for a general review.
Article
Acute myeloid leukemia (AML) occurs as the result of malignant transformation in a hematopoietic progenitor cell, which proliferates to form an accumulation of AML blasts. Only a minority of these AML cells are capable of proliferation in vitro, suggesting that AML cells may be organized in a hierarchy, with only the most primitive of these cells capable of maintaining the leukemic clone. To further investigate this hypothesis, we have evaluated a strategy for purifying these primitive cells based on surface antigen expression. As an in vitro endpoint, we have determined the phenotype of AML progenitor cells which are capable of producing AML colony-forming cells (CFU) for up to 8 weeks in suspension culture (SC) and compared the phenotype with that of cells which reproduce AML in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. AML cells were fluorescence-activated cell sorted (FACS) for coexpression of CD34 and CD71, CD38, and/or HLA-DR and the subfractions were assayed in vitro and in vivo at various cell doses to estimate purification. While the majority of primary AML CFU lacked expression of CD34, most cells capable of producing CFU after 2 to 8 weeks in SC were CD34+/CD71−. HLA-DR expression was heterogeneous on cells producing CFU after 2 to 4 weeks. However, after 6 to 8 weeks in SC, the majority of CFU were derived from CD34+/HLA-DR− cells. Similarly, the majority of cells capable of long-term CFU production from SC were CD34+/CD38−. Most cells that were capable of engrafting NOD/SCID mice were also CD34+/CD71− and CD34+/HLA-DR−. Engraftment was not achieved with CD34+/CD71+ or HLA-DR+subfractions, however, in two patients, both the CD34+and CD34− subfractions were capable of engrafting the NOD/SCID mice. A three-color sorting strategy combining these antigens allowed approximately a 2-log purification of these NOD/SCID leukemia initiating cells, with engraftment achieved using as few as 400 cells in one experiment. Phenotyping studies suggest even higher purification could be achieved by combining lack of CD38 expression with the CD34+/CD71− or CD34+/HLA DR− phenotype. These results suggest that most AML cells capable of long-term proliferation in vitro and in vivo share the CD34+/CD71−/HLA-DR− phenotype with normal stem cells. Our data suggests that in this group of patients the leukemic transformation has occurred in a primitive progenitor, as defined by phenotype, with some degree of subsequent differentiation as defined by functional assays.
Article
Analysis of the mitogenic activity of interleukin-3 (IL-3), Steel factor (SF ), and flt-3 ligand (FL) on acute myelogenous leukemia (AML) blasts using the short-term endpoints of proliferation in 3H-thymidine (3H-Tdr) incorporation assays or methylcellulose cultures (colony assays) showed that greater than 90% of samples contained cells that were responsive to one or more of these cytokines. With this information, culture conditions that were known to support normal long-term culture-initiating cells (LTC-IC) were tested, with or without supplements of one or more of these three growth factors, for their ability to support primitive progenitors from 10 cell samples from patients with AML. In all cases cytogenetically abnormal colony forming cells (CFC) were detected after 5 weeks when AML peripheral blood or marrow cells were cocultured on preestablished, normal human marrow feeders (HMF ) and/or Sl/Sl mouse fibroblast feeders and the number of CFC detected in these 5-week-old LTC maintained a linear relationship to the number of input AML cells. Limiting dilution analysis, performed on 6 of the 10 samples, showed the frequency of AML cells initiating LTC (AML LTC-IC) to be 5- to 300-fold lower than the frequency of AML-CFC in the same cell sample, whereas the average number of CFC produced per LTC-IC varied from 1 to 13. Surprisingly, in each case the concentration of cytogenetically normal LTC-IC detected in AML patient blood was at least 10-fold higher than that previously observed in the blood of normal individuals. “Mixed” mouse fibroblast feeders engineered to produce human G-CSF, IL-3, and SF did not enhance detection of AML LTC-IC but did increase the output of cytogenetically normal CFC from LTC of 3 of 4 patient samples. Supplementation of AML LTC with IL-3 and exogenously provided SF and/or FL increased the output of AML-CFC from 5-week-old LTC by greater than or equal to twofold with 5 of 9 patient samples, whereas in one case exogenous addition of FL reduced the output of malignant CFC from LTC. These studies show that conditions that support normal LTC-IC also allow a functionally analogous but rare AML progenitor cell type to be detected. In addition, differences in the responses of normal and leukemic cells to various cytokines active on normal LTC-IC were revealed. Further analysis of these differences may enhance our understanding of leukemogenesis and lead to observations that could be exploited therapeutically.
Article
Somatic mutations of the receptor tyrosine kinase Flt3 consisting of internal tandem duplications (ITD) occur in 20% of patients with acute myeloid leukemia. They are associated with a poor prognosis of the disease. In this study, we characterized the oncogenic potential and signaling properties of Flt3 mutations. We constructed chimeric molecules that consisted of the murine Flt3 backbone and a 510-base pair human Flt3 fragment, which contained either 4 different ITD mutants or the wild-type coding sequence. Flt3 isoforms containing ITD mutations (Flt3-ITD) induced factor-independent growth and resistance to radiation-induced apoptosis in 32D cells. Cells containing Flt3-ITD, but not those containing wild-type Flt3 (Flt3-WT), formed colonies in methylcellulose. Injection of 32D/Flt3-ITD induced rapid development of a leukemia-type disease in syngeneic mice. Flt3-ITD mutations exhibited constitutive autophosphorylation of the immature form of the Flt3 receptor. Analysis of the involved signal transduction pathways revealed that Flt3-ITD only slightly activated the MAP kinases Erk1 and 2 and the protein kinase B (Akt) in the absence of ligand and retained ligand-induced activation of these enzymes. However, Flt3-ITD led to strong factor-independent activation of STAT5. The relative importance of the STAT5 and Ras pathways for ITD-induced colony formation was assessed by transfection of dominant negative (dn) forms of these proteins: transfection of dnSTAT5 inhibited colony formation by 50%. Despite its weak constitutive activation by Flt3-ITD, dnRas also strongly inhibited Flt3-ITD–mediated colony formation. Taken together, Flt3-ITD mutations induce factor-independent growth and leukemogenesis of 32D cells that are mediated by the Ras and STAT5 pathways.
Article
Despite the usual uniform and primitive appearance of cells derived from the leukemic clone in most patients with acute myeloid leukemia (AML), there is considerable heterogeneity among leukemic blasts, particularly with respect to their capacity to proliferate and/or self renew. We have assessed whether these differences in proliferative potential are correlated with the phenotypic changes that characterize normal hematopoiesis, which might suggest an analogous hierarchy of AML progenitors. We have used the ability of primitive AML cells to persist or produce blast colony forming cells (CFU-blast) detected after 2 to 8 weeks in the presence of growth factors in suspension cultures (SC) termed SC-initiating cells (IC), or with stroma in long-term cultures (LTC-IC) as a quantitative assay for a cell that may have primitive characteristics. This SC assay is linear, cell concentration independent, and the frequency of SC-IC by limiting dilution analysis is lower than primary CFU-blast. The average output of CFU-blast after 2 to 8 weeks by individual SC-IC varied between 2 and more than 100 in individual patients. Leukemic blasts were sorted based on their expression of antigens previously found useful to characterize normal progenitor differentiation, and analyzed for the percentage of CFU- blast SC-IC, and leukemic LTC-IC within each fraction. All of these progenitor types were heterogeneous in their expression of CD45RA and CD33, but expressed uniformly low levels of CD15 and differed from normal primitive progenitors in their high expression of HLA-DR. CFU- blast had a significantly higher expression of CD71 and CD38 as compared with SC-IC or leukemic LTC-IC. In patients with CD34+ blasts, the majority of their SC-IC at 4 weeks were CD34+/CD38-; however, patients with CD34- blasts had at least some CD34- progenitors. These results show that while heterogeneity exists between patients, it is possible to physically separate subpopulations of AML cells with different proliferative potentials. It also provides some support for the concept that quantitation of leukemic cells capable of producing CFU-blast for 4 weeks or more in vitro measures a less frequent leukemic progenitor with higher proliferative potential that may be the only relevant cell for maintaining the leukemic clone in vivo.
Article
Chemotherapeutic agents simultaneously induce transcription factors p53 and NFκB. p53 induction can activate an apoptotic program, and resistance to chemotherapy correlates with the loss of a functional p53 pathway. By contrast, NFκB prevents apoptosis in response to chemotherapeutic agents. We have analyzed the p53 response in IKK1/2−/− MEFs, which lack detectable NFκB activity. Compared to WT fibroblasts, IKK1/2−/− fibroblasts showed increased cell death and p53 induction in response to the chemotherapeutic agent, doxorubicin. Reconstitution of IKK2, but not IKK1, increased Mdm2 levels and decreased doxorubicin-induced p53 stabilization and cell death. IKK2-mediated effects required its kinase function and were abrogated by coexpression of the dominant negative IκBαM, implying a role for NFκB in blocking chemotherapy-induced p53 and cell death.
Article
Previous studies indicate that human acute myelogenous leukemia (AML) arises from a rare population of leukemic stem cells. Cells of this nature can initiate and maintain leukemic cell growth in both long-term cultures and nonobese diabetic/severe combined immune-deficient mice. To characterize the biology of primitive AML cells, gene expression screens were performed with 7 primary AML and 3 normal specimens. For each sample, stem cell populations (CD34+/CD38−) were isolated and used to synthesize radiolabeled complementary DNA (cDNA). AML vs normal probes were then hybridized to cDNA arrays containing genes related to cancer and apoptosis. Of approximately 1400 genes analyzed, 2 tumor-suppressor genes were identified that were overexpressed in all 7 of the AML CD34+/CD38−cell populations: death-associated protein kinase and interferon regulatory factor 1. Expression of each gene was confirmed by reverse-transcription polymerase chain reaction and immunoblot analysis. It is proposed that tumor-suppressor proteins play a role in the biology of primitive AML cells.
Article
Summary Chemotherapeutic agents simultaneously induce transcription factors p53 and NFB. p53 induction can activate an apo- ptotic program, and resistance to chemotherapy correlates with the loss of a functional p53 pathway. By contrast, NFB prevents apoptosis in response to chemotherapeutic agents. We have analyzed the p53 response in IKK1/2 / MEFs, which lack detectable NFB activity. Compared to WT fibroblasts, IKK1/2 / fibroblasts showed increased cell death and p53 induction in response to the chemotherapeutic agent, doxorubicin. Reconstitution of IKK2, but not IKK1, increased Mdm2 levels and decreased doxorubicin-induced p53 stabilization and cell death. IKK2-mediated effects required its kinase function and were abrogated by coexpression of the dominant negative IBM, implying a role for NFB in blocking chemotherapy-induced p53 and cell death.
Article
The 26S proteasome is a non-lysosomal multicatalytic protease complex for degrading intracellular proteins by ATP/ubiquitin-dependent proteolysis. Tightly ordered proteasomal degradation of proteins critical for cell cycle control implies a role of the proteasome in maintaining cell proliferation and cell survival. In this study, we demonstrate that cell-permeable proteasome inhibitors, lactacystin, benzyloxycarbonyl(Z)-leucyl-leucyl-leucinal (ZLLLal; MG-132) and4-hydroxy-5-iodo-3-nitrophenylacetyl-leucyl-leucyl-leucine vinyl sulfone (NLVS), induce apoptosis abundantly in p53-defective leukemic cell lines CCRF-CEM, U937 and K562 as well as in myelogenic and lymphatic leukemic cells obtained from adult individuals with relapsed acute leukemias. Leukemic cell apoptosis induced by the proteasome inhibitors was dependent on activation of caspase-3 and related caspase family proteases, because caspase-3 inhibitor N-acetyl-l-aspartyl-l-glutamyl-l-valyl-l-aspartal (Ac-DEVD-cho) and, more effectively, the general caspase-inhibitor N-benzyloxycarbonyl-l-valyl-l-alanyl-l-aspartate fluoromethylketone (Z-VAD-fmk) were capable of blocking apoptosis induced by lactacystin, ZLLLal or NLVS. Induction of apoptosis by lactacystin or ZLLLal was accompanied by cell cycle arrest at G2/M phase and by accumulation and stabilization of cyclin-dependent kinase inhibitor p21WAF1/Cip and tumor suppressor protein p53. A role of p53 in mediating apoptosis or induction of p21WAF1/Cip1 was ruled out since CCRF-CEM and U937 cells express non-functional mutant p53, and K562 cells lack expression of p53. Viability and hematopoietic outgrowth of human CD34+ progenitor cells treated with lactacystin were slightly reduced, whereas treatment of CD34+ cells with ZLLLal or the cytostatic drugs doxorubicin and gemcitabine resulted in markedly reduced viability and hematopoietic outgrowth. These results demonstrate a basic role of the proteasome in maintaining survival of human leukemic cells, and may define cell-permeable proteasome inhibitors as potently anti-leukemic agents which exhibit a moderate hematopoietic toxicity in vitro.