Multiple Functions of The 37-67-kd Laminin Receptor

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

© The American Society of Gene & Cell Therapy

original article

Multiple Functions of the 37/67-kd Laminin


Receptor Make It a Suitable Target for Novel
Cancer Gene Therapy
Jonathan Scheiman1, Jen-Chieh Tseng1, Yun Zheng1 and Daniel Meruelo1
NYU Cancer Institute and the NYU Gene Therapy Center, NYU School of Medicine, New York, New York, USA
1

The 37/67-kd laminin receptor, LAMR, is a ­multifunctional cancer7–12 where as an extracellular matrix molecule, it is important
protein that associates with the 40S ribosomal sub- for tumor-cell migration,13 invasion, and angiogenesis.14 Further,
unit and also localizes to the cell membrane to inter- LAMR can remodel laminin-1 to alter tumor-cell gene expression
act with the extracellular matrix. LAMR is overexpressed and increase tumor aggressiveness,15 as well as alter intracellular
in many types of cancer, playing important roles in signaling pathways.16 LAMR is therefore considered a useful prog-
tumor-cell migration and invasion. Here, we show that nostic marker for determining the severity of tumors.17
LAMR is also vital for tumor-cell proliferation, survival, The 67-kd LAMR was first discovered by three independent
and protein translation. Small-interfering RNA (siRNA)– laboratories in 1983 (refs. 18–20) through its ability to bind to and
mediated reduction in expression of LAMR leads to G1 be isolated by laminin sepharose. Its gene, however, was found to
phase cell-cycle arrest in vitro by altering cyclins A2/B1, encode a protein of only 37 kd. The discrepancy between these two
cyclin-dependent kinases (CDKs) 1/2, Survivin, and p21 molecular weights was later resolved by showing that the 37-kd
expression levels. In vivo, reduction in LAMR expression gene product serves as a monomeric precursor to a 67-kd ­dimer.21
results in inhibition of HT1080 cells to develop tumors. The exact composition of the 67-kd dimer and the process by
We also found that LAMR’s ribosomal functions are criti- which it is formed remains obscure as evidence supports both a
cal for translation as reduction in LAMR expression leads homo22 and a heterodimer.23,24 LAMR was shown to be acylated by
to a dramatic decrease in newly synthesized proteins. three fatty acids—palmitate, stearate, and oleate22—and fatty acid
Further, cells with lower expression of LAMR have fewer synthesis is required for 67-kd LAMR formation.23 Beyond this not
40S subunits and 80S monosomes, causing an increase much is known about what regulates the dimerization process.
in free 60S ribosomal subunits. These results indicate The 37-kd LAMR monomer is not without its own intrigue.
that LAMR is able to regulate tumor development in It is a highly conserved ribosomal protein that acquired its extra-
many ways; further enhancing its potential as a target cellular matrix functions during evolution.25 The 37-kd LAMR,
also known as p40, is ubiquitously expressed in many organisms
for gene therapy. To test this, we developed a novel
as a 40S ribosome–associated protein. The mammalian sequence
Sindbis/Lenti pseudotype vector carrying short-hairpin
has homologues in many different organisms including bacteria,
RNA (shRNA) designed against lamr. This pseudotype
yeast, plant (reviewed in ref. 25). The 37-kd LAMR/p40 has been
vector effectively reduces LAMR expression and specifi-
shown to be polysome associated in both yeast26 and plant.27 The
cally targets tumors in vivo. Treatment of tumor-bearing
ribosomal functions of 37-kd LAMR are essential for cell viability
severe combine immunodeficient (SCID) mice with this
in yeast; 37-kd LAMR/p40 is required for processing 20S to 18S
pseudotype vector significantly inhibits tumor growth.
ribosomal RNA and thus for maturation of the 40S ribosomal sub-
Thus, we show that LAMR can be used as a target in
unit and 80S monosome assembly.28 The 37-kd LAMR/p40 is also
novel therapy for tumor reduction and elimination. required for HeLa29 and Hep3b30 cell viability, although in HeLa
Received 11 October 2009; accepted 28 July 2009; published online cells loss of 37-kd LAMR/p40 reportedly did not inhibit protein
1 September 2009. doi:10.1038/mt.2009.199 translation and therefore was thought not to be the cause of cell
death. LAMR has also been shown to localize to the nucleus and
Introduction interact with histones H2A, H2B, and H4 (ref. 31). Thus nuclear
The 37/67-kd laminin receptor (LAMR) plays a role in many path- localization/functions of LAMR may play a critical role in cell
ological processes. It serves as a cell-surface receptor for prions,1 viability. However, the fact that LAMR is polysome associated in
cytotoxic necrotizing factor-1 expressing Escherichia coli K1 (ref. 2) mammalian cells32 suggests that it retains some ribosomal func-
and numerous viruses, including Sindbis,3 dengue,4 Venezuelan tion. The importance of 37-kd LAMR as a ribosomal protein and
equine encephalitis,5 and adeno-associated virus subtypes 2, 3, how it maintains cell viability in mammalian cells still needs to be
8, and 9 (ref. 6). LAMR is also over expressed in many types of thoroughly examined and is something we wanted to investigate.

Correspondence: Daniel Meruelo, NYU Cancer Institute and the NYU Gene Therapy Center, NYU School of Medicine, 550 First Avenue, New York,
New York 10016, USA. E-mail: merued01@med.nyu.edu

Molecular Therapy vol. 18 no. 1, 63–74 jan. 2010 63


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

Considering the large number of cellular processes that quantitative real-time PCR and western blot analysis (Figure 1b
LAMR governs and the overwhelming evidence for its involve- left and right, respectively). In our system, we are able to achieve
ment in pathology, further understanding of its functionality >90% knock down of LAMR expression at both the mRNA and
is important for developing it as a target for new therapeutic protein levels.
strategies. LAMR is already a target for prion disease therapy.33 We observed that siLAMR-transfected cells grow slower than
Also, many studies have focused on the extracellular functions control-transfected cells (Figure 1c, left). Cell proliferation was
of LAMR for cancer therapeutic purposes. For instance, using therefore assessed. Equal numbers of nontransfected, siLAMR-,
various anti-LAMR agents impede tumor-cell invasion in vitro34 and siGLO-transfected cells were seeded 2 days after transfection
and antibodies that specifically bind LAMR can inhibit tumor and allowed to grow for 3 days after seeding. As Figure 1c shows
metastasis in vivo.35 Currently, it is not known what role the 37-kd (right), there was a twofold reduction in siLAMR-transfected
monomer plays in tumor development. Targeting intracellular cell number compared to control cells 1 day after seeding cells,
and ribosomal functions of 37-kd LAMR might be as effective as ­followed by a 5- and 6.5-fold reduction 2 and 3 days after seeding,
inhibiting extracellular functions although has been explored to a respectively. Thus, cells with reduced expression of LAMR have a
lesser extent. Therefore, we seek to examine how the 37-kd LAMR lower proliferation rate. This phenomenon was also observed with
monomer may effect tumor development using small-interfering 293, HeLa, and HepG2 cells (Figure 1d) suggesting that LAMR
RNA (siRNA) to silence lamr expression and to assess whether we plays an important role in cellular growth rate.
can improve upon current strategies that target LAMR solely as
an extracellular molecule. Silencing total lamr expression might Reduction in LAMR expression results in G1 phase
inhibit multiple LAMR functions and therefore more effectively cell-cycle arrest
address questions about cell viability as well as be more efficient Cell-cycle profiles of nontransfected, siLAMR-, and siGLO-trans-
in treating tumors in vivo. In this study, we focus exclusively on fected cells were compared 3–6 days after transfection by stain-
the expression of the 37-kd monomer, and therefore refer to the ing with propidium iodide and analyzing by fluorescent activated
monomer whenever we mention LAMR. cell sorting. Profiles are summarized in Figure 2a. At 4 days after
Targeting genes for cancer gene therapy in vivo, as we seek transfection, ~40% of both nontransfected and siGLO cells were in
to do with LAMR, has hitherto been greatly limited by lack of an G1 phase compared to about 55% for the siLAMR-transfected cells
efficient delivery system for tumor targeting. A vector system is (Figure 2a, left panel) and ~40% of the control cells are in S phase
needed to achieve specific and efficient tumor targeting in living compared to about 25% of siLAMR-transfected cells (Figure 2a,
animals. Several vectors based on lentivirus (e.g., human immu- right panel). The disparity becomes even greater at 5 days after
nodeficiency virus) have been developed to deliver foreign DNA transfection (Figure 2b) when 30% of nontransfected and 28% of
into cells without significant safety concerns. The core of lentivi- siGLO-transfected cells were in the G1 phase compared to 65%
rus can accommodate a wide range of glycoproteins from other of siLAMR-transfected cells. Furthermore, 55% of nontransfected
viruses, providing a relatively flexible choice of viral glycoprotein and 49% of siGLO-transfected cells were in S phase compared to
for specific targeting. Mentioned earlier, LAMR is a cellular recep- only 16% of the siLAMR-transfected cells. These results indicate
tor for Sindbis virus. We have previously developed Sindbis viral that cells with reduced levels of LAMR undergo cell-cycle arrest
vectors as therapeutic agents to specifically target and kill tumor in the G1 phase. The arrest is only transient, however, as the cell-
cells and have also shown that Sindbis’ tumor-targeting ability cycle profiles of siLAMR-transfected cells are similar to those of
is mediated in part by LAMR.36 Thus, not only is LAMR itself a the control cells 6 days after transfection as shown in Figure 2a.
potential focus for cancer therapy, it also is a means of targeting
tumors with Sindbis viral vectors. Here, we explore the use of a LAMR expression affects expression levels of cell
Sindbis/Lenti pseudotype vector, carrying a short-hairpin RNA cycle–related genes
(shRNA) cassette, to target tumors cells in vivo and effectively Because siLAMR-transfected cells undergo cell-cycle arrest,
reduce LAMR expression for cancer gene therapy. we investigated whether any cell cycle–related gene ­expression
is altered. Using the cell cycle RT2Profiler PCR array from
Results SuperArray (Frederick, MD), the expression levels of 84 different
Knocking down LAMR expression inhibits genes were compared between siLAMR- and siGLO-transfected
cell proliferation cells. Several genes were found whose expression was altered in
To determine the role that LAMR plays in tumor-associated func- siLAMR-transfected cells in a manner consistent with G1 arrest
tions, we transiently knocked down its expression in the HT1080 (data not shown). These genes, validated by quantitative real-time
human fibrosarcoma cell line by transfection with a predesigned PCR at 3 and 4 days after transfection (Figure 3a left and right,
siRNA pool targeting human LAMR (siLAMR). This pool consists respectively), included cyclins A/B, cyclin-dependent kinases
of four individual oligonucleotides that target different regions (CDKs) 1/2, E2F1, and p21.
of the human LAMR sequence (Supplementary Figure S1). At the mRNA level, p21, a cyclin-dependent kinase inhibitor
Fluorescently labeled nontargeting control siRNA (siGLO) was and tumor suppressor gene that inhibits cell-cycle progression
used as a control. Fluorescent activated cell sorting analysis shows at the G1/S check point, was upregulated in siLAMR-transfected
that ~97% of HT1080 cells are transfected with siGLO (Figure 1a), cells 15-fold 3 days after transfection and eightfold 4 days after
indicating a high level of siRNA transfection efficiency. We ana- transfection (Figure 3a left and right, respectively). In contrast,
lyzed the efficiency of LAMR knock down via siLAMR by both cyclin A2, expressed in S/G2, and cyclin B1, expressed in G2/M

64 www.moleculartherapy.org vol. 18 no. 1 jan. 2010


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

a 100
siGLO
80

% of Max
60
97.2
40 Non-
transfected siLAMR siGLO

20 LAMR

0
100 101 102 10
3
104 β-Actin
FL2-H

b LAMR mRNA expression LAMR protein expression

Nontransfected Nontransfected
2.5
1.5 siLAMR
siLAMR
siGLO 2.0 siGLO

1.0

Fold change
Fold change

1.5

1.0
0.5
0.5

*** ***
0.0 0.0

c Nontransfected siGLO siLAMR

HT1080
7,500,000
Day 1 Nontransfected
Relative light units

siLAMR
5,000,000 siGLO

Day 2 2,500,000

0
1 2 3
Day 3 Time (days)

d 293 HeLa HepG2


Nontransfected 30,000,000 30,000,000
100,000,000
siLAMR
Relative light units

siGLO
75,000,000 20,000,000 20,000,000

50,000,000
10,000,000 10,000,000
25,000,000

0 0 0
1 2 3 4 5 1 2 3 4 1 2 3 4 5
Time (days)

Figure 1 Reduction in laminin receptor (LAMR) expression inhibits cell proliferation. (a) HT1080 cells transfected with the fluorescently labeled
nontargeting small-interfering RNA (siRNA) control, siGLO, were analyzed with a FACSCaliber machine and gated according to nontransfected cells
(dotted line). Approximately 97% of siGLO-transfected cells (solid line) were positive for siGLO, as determined by Flowjo 8.2 software (Tree Star).
Fluorescent activated cell sorting analysis was performed 1 day after transfection. (b) HT1080 cells transfected with an siRNA pool targeting LAMR
(siLAMR) were harvested for RNA 3 days after transfection and protein extraction 4 days after transfection. Quantitative real-time PCR was used to
check LAMR mRNA expression (left) and western blot analysis was used to check LAMR protein levels (top right), using glyceraldehyde-3-phosphate
dehydrogenase (GAPDH) and β-actin as internal controls, respectively. Western blot results were quantified and graphed (bottom right). Statistical
analysis was performed using a standard Student’s t-test to generate P values. All P values are two-tailed (***P < 0.0001). (c) Images of nontransfected,
siLAMR-, and siGLO-transfected cells were taken 1–3 days after seeding for proliferation assay (3–5 days after transfection, respectively) (left). Cell pro-
liferation was measured for HT1080 nontransfected, siLAMR-, and siGLO-transfected cells at the same time points (right). (d) Similar cell ­proliferation
assays were performed for 293, HeLa, and HepG2 cell lines.

phases were downregulated 90% at both time points. CDKs1 and when comparing siLAMR-transfected cells to siGLO-transfected
2, which promote cell-cycle progression, are both reduced ~60% cells. At the mRNA level, there was generally no significant dif-
3 days after transfection (Figure 3a, left). In addition, the E2F1 ference between nontransfected and siGLO-transfected cells
transcription factor, expressed during S phase, was downregu- in the genes analyzed. In the few exceptions when there was,
lated ~60% 3 days after transfection (Figure 3a, left). The statis- it only enhanced the disparity between siLAMR- and siGLO-
tical significance of each gene was evaluated to generate P values ­transfected cells.

Molecular Therapy vol. 18 no. 1 jan. 2010 65


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

a Percentage of HT1080 cells in Percentage of HT1080 cells in


G1 phase S phase
75 75
Nontransfected
siLAMR
% 50 50 siGLO

25 25

0 0
3 4 5 6 3 4 5 6
Time (days after transfection)

b Nontransfected siLAMR siGLO

800 800 800

600 600 600


Cell number

G1 G1 G1
400 30% 400 65% 400 28%
S 55% S 16% S 49%
200 200 200

0 0 0
0 30 60 90 120 150 0 30 60 90 120 150 30 60 90 120 150
Channels

Figure 2 siLAMR-transfected cells undergo cell-cycle arrest in the G1 phase. (a) Summary of cell-cycle profiles for HT1080 siRNA-transfected
cells. Cells were stained with propidium iodide and analyzed by fluorescent activated cell sorting. Percentage of cells in G1 phase (left) and S
phase (right) are shown 3–6 days after transfection. (b) Cell-cycle profiles 5 days after transfection with siRNA. Percentage of cells in G1 and S phase
are indicated (arrows). siGLO, fluorescently labeled nontargeting siRNA control; siLAMR, siRNA pool targeting human laminin receptor; siRNA, small
interferin RNA.

Protein levels of these gene products were measured 4 days LAMR expression regulates translation
after transfection. As Figure 3b (left) shows, knock down of Because LAMR is a ribosomal protein, we wanted to determine
LAMR resulted in a dramatic decrease in cyclins A/B, and whether it plays a role in translation in HT1080 cells. Novo pro-
CDKs 1/2 at the protein level. On the other hand, p21, barely tein synthesis by nontransfected, siLAMR- and siGLO-trans-
expressed in control cells, was upregulated in siLAMR-trans- fected cells was measured 2–4 and 7 days after transfection by
fected cells. β-Actin was used as a loading control and shows pulsing cells with 35S-methionine for 2 hours and chasing for an
that equal amounts of protein were loaded. Western blots are additional 90 minutes. Equal amounts of protein were loaded for
representative of at least three transfections. Protein ­expression sodium dodecyl sulfate–polyacrylamide gel electrophoresis and
levels were quantified and statistically analyzed comparing siL- used to detect newly synthesized proteins. As Figure 4a shows,
AMR- and siGLO-transfected cells (Figure 3b, right). These translation was markedly inhibited in siLAMR-transfected cells
results indicate that reduction in LAMR expression alters the at all time points, indicating that LAMR expression is critical
expression of genes and proteins that are important for cell- for this process. In addition, treating siLAMR-transfected cells
cycle progression. with trypsin and reseeding them the day before labeling had no
Another gene shown to be downregulated in the Cell-cycle ­additional effect on translation inhibition, indicating that this
SuperArray and validated through real-time PCR was Survivin. process is not regulated by extracellular functions of LAMR
An antiapoptotic protein overexpressed in many types of cancer, important for cell adhesion.
Survivin was reduced 90% both at the mRNA level and protein LAMR was previously shown to be required for 40S ribo-
level in siLAMR-transfected cells (Figure 3a,b). At the protein somal subunit maturation and 80S monosome assembly in
level, there is also a significant reduction of Survivin in siGLO- yeast.28 To analyze ribosomal integrity in siLAMR- and siG-
transfected cells compared to nontransfected cells, which may be LO-transfected cells, sucrose-gradient ultracentrifugation was
due to the transfection procedure. However, the fact that Survivin used. Equal numbers of cells were lysed and separated through
is reduced at the mRNA level only in siLAMR-transfected cells 10–50% linear sucrose gradients 2 and 3 days after transfection.
and almost completely absent in siLAMR-transfected cells at the A total of 24 fractions of equal volume were collected from the
protein level suggests that reduction of Survivin expression is a top of each gradient and their optical density was measured
specific effect of LAMR knock down. In addition, it also suggests at A260 nm to generate ribosomal profiles (Figure 4b). Two days
that tumor cells with reduced levels of LAMR may also be more after transfection, there was a modest reduction in the 80S
susceptible to cell death. monosome peak in siLAMR-transfected cells (Figure 4b, left).

66 www.moleculartherapy.org vol. 18 no. 1 jan. 2010


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

a HT1080 siLAMR HT1080 siLAMR


Cell cycle gene expression Cell cycle gene expression
(3 days) (4 days)
Nontransfected
15.0 9
siLAMR
12.5 siGLO 7

10.0 5
Fold change

1 1

0 0
R

A2

B1

K1

K2

21

A2

B1

K2

F1

in

21
2F

vi
AM

iv
*p

*p
E2
D

D
vi
lin

lin

lin

lin

rv
LA
*E
*C

*C
ur
*L

Su
**
yc

yc

yc

yc
**
*S
**
*C

*C

**
**

**
Gene

b Nontransfected siLAMR siGLO


LAMR HT1080 siLAMR
(37 kd) Cell cycle protein expression

Cyclin A2 Nontransfected
5
(50 kd)
siLAMR
4
Cyclin B1 siGLO
(50 kd) 3

CDK1 2
Fold change

(34 kd) 1.5

CDK2
(34 kd) 1.0

Survivin
0.5
(15 kd)

p21 0.0
(21 kd)
R

A2

B1

K1

K2

in

21
M

iv

*p
D

D
lin

lin

rv
LA

*C

**
Su
yc

yc

**
**

**

β-Actin
*C

**
**
**

(42 kd) Protein

Figure 3 siLAMR alters the expression of cell cycle–related genes and proteins. (a) Quantitative real-time PCR validation of cell cycle–related
genes found to be altered in HT1080 siLAMR–transfected cells 3 days (left) and 4 days (right) after transfection. Glyceraldehyde-3-phosphate dehy-
drogenase was used as an internal control. Results are indicative of at least two separate transfections. (b) Western blot analysis of cell cycle–related
proteins 4 days after transfection. Protein (20 μg) was loaded. β-Actin was used as a control. Western blots are indicative of three separate trans-
fections (left). Observed molecular weights of each protein are indicated in parentheses. Protein expression levels were quantified and statistically
­analyzed (right) (*P < 0.05. **P < 0.001. ***P < 0.0001). CDK, cyclin-dependent kinase; siGLO, fluorescently labeled nontargeting siRNA control;
siLAMR, siRNA pool targeting human LAMR; siRNA, small interferin RNA.

Three days after transfection, the 80S monosome peak was LAMR is critical for tumor growth in vivo
dramatically reduced in siLAMR-transfected cells, which was To further examine the effects of knocking down LAMR expres-
accompanied by a reduction in the 40S peak and an increase in sion, we assessed the ability of cells with reduced levels of LAMR
the 60S peak (Figure 4b, right). to grow tumors in vivo. An HT1080 cell line was generated
Fractions 1–12 from the 3-day time point were further ana- that stably expresses the firefly luciferase gene (HT1080 FLUC)
lyzed by western blot, probing for LAMR, 40S ribosomal pro- ­permitting it to be visualized and quantified in vivo with use of
tein S6, and 60S ribosomal protein L7a. As Figure 4c shows, an in vivo imaging system (IVIS). A volume of 1 × 106 nontrans-
­reduction in LAMR resulted in a shift of L7a from fractions 9 fected, siLAMR-, and siGLO-transfected HT1080 FLUC cells
and 10, which corresponds with the 80S monosome peak in were injected subcutaneously into mice 2 days after transfection
the siGLO-transfected ribosomal profile, to fractions 7 and 8, (five mice were injected separately for each group). In addition,
­corresponding to the 60S subunit fractions. S6 was also reduced cells were reseeded in tissue culture and harvested for RNA 4
in fractions 9 and 10. These blots therefore confirmed the ribo- days after transfection. siLAMR-transfected cells had over an 80%
somal profiles, and show that LAMR is crucial for 40S matu- reduction of LAMR mRNA compared to siGLO-transfected cells
ration and 80S ­monosome assembly in HT1080 cells. Without (Figure 5a).
LAMR, there is a reduction in 40S subunits, which results in Mice were imaged 3, 10, 17, and 24 days after injection (5, 12,
an increase of unassociated 60S ribosomal subunits, less 80S 19, and 26 days after transfection, respectively) to monitor tumor
monosomes, and a dramatic ­reduction in translation as men- development of HT1080 FLUC fibrosarcoma cells (Figure 5b,
tioned above. left). Tumor-cell fluorescence, which stayed localized to the sight

Molecular Therapy vol. 18 no. 1 jan. 2010 67


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

a 2 days 3 days

35
S-methionine

LAMR

β-Actin
Non- siLAMR siGLO Non- siLAMR siLAMR siGLO
transfected transfected Trypsin

4 days 7 days

35
S-methionine

LAMR
β-Actin
Non- siLAMR siLAMR siGLO Non- siLAMR siLAMR siGLO
transfected Trypsin transfected Trypsin

b 1.4 2 days
1.2
3 days

1.2 1 siGLO
1 siLAMR
0.8
0.8 40S 60S 80S 40S 60S 80S
A260

0.6
0.6
0.4
0.4

0.2 0.2

0 0
0 5 10 15 20 25 0 5 10 15 20 25
Fraction

c siGLO siLAMR
LAMR

L7a

S6
4 5 6 7 8 9 10 4 5 6 7 8 9 10
Fraction

Figure 4 Laminin receptor (LAMR) expression regulates translation. (a) Nontransfected, siLAMR-, and siGLO-transfected cells were metabolically
labeled with 35S-methionine, 2–4, and 7 days after transfection. Labeling of cells was performed in triplicate. Protein (20 μg) was loaded for sodium
dodecyl sulfate–polyacrylamide gel electrophoresis to detect newly synthesized proteins. siLAMR-transfected cells were either allowed to remain
attached to dishes or treated with trypsin the day before labeling and then reseeded (siLAMR trypsin). Western blot analysis was used to check for
LAMR and β-actin expression. (b) An equal number of siLAMR- and siGLO-transfected cells were collected and lysed 2 and 3 days after transfection.
Cell lysates were separated on a 10–50% linear sucrose gradient and collected in 24 fractions of equal volume. The optical density of each fraction
was measured at A260 nm to generate ribosomal profiles. Solid lines represent siLAMR-transfected profiles, dashed lines represent siGLO-transfected
profiles. Arrows indicate 40S, 60S, and 80S peaks. (c) Thirty-two microliters of fractions 1–12 from the siLAMR and siGLO ribosomal gradients, 3 days
after transfection, were loaded for western blot analysis. Membranes were blotted for LAMR, L7a, and S6. siGLO, fluorescently labeled nontargeting
siRNA control; siLAMR, siRNA pool targeting human LAMR; siRNA, small interferin RNA.

of injection, was then quantified (Figure 5b, right). At 3 days after later time points, siLAMR-transfected tumors show a decrease in
injection, control cells had an average fluorescence about two- luciferase signal intensity and by 17 days the siLAMR-transfected
fold higher than siLAMR-transfected cells, which increased to tumor cells can no longer be detected (Figure 5b). This indicates
1,000-fold after 10 days, consistent with the higher proliferation that siLAMR-transfected HT1080 cells have an impaired ability to
rate of control versus siLAMR-transfected cells seen in vitro. At grow in vivo. In contrast to the other experimental groups, animals

68 www.moleculartherapy.org vol. 18 no. 1 jan. 2010


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

a HT1080 FLUC
LAMR mRNA expression
1.0 Nontransfected
siLAMR
siGLO

Fold change
0.5

0.0 ***

b Day 3 10 17 24
Nontransfected

60,000 HT1080 tumor growth curves

50,000 5.0 × 107

40,000 4.0 × 107

Photon counts
30,000 3.0 × 107
siLAMR

20,000 Nontransfected (n = 5)
2.0 × 107
10,000 siLAMR (n = 5)
7
1.0 × 10
siGLO (n = 5)
Counts
0
Color Bar 0 10 20 30 40 50 60
Min = 600 Days after inoculation
Max = 60,000
siGLO

Figure 5 LAMR is critical for HT1080 tumor-cell growth and survival in vivo. (a) HT 1080 FLUC nontransfected, siLAMR-, and siGLO-transfected
cells were harvested for RNA extraction 4 days after transfection. Quantitative real-time PCR was performed to determine LAMR mRNA expression
levels. Glyceraldehyde-3-phosphate dehydrogenase was used as an internal control. (***P < 0.0001). (b) Severe combine immunodeficient mice
were injected subcutaneously with HT1080 FLUC nontransfected, siLAMR-, and siGLO-transfected cells and allowed to grow tumors. Luciferase signal
intensity was measured 3, 10, 17, and 24 days after injection with use of in vivo imaging system (left) and quantified (right). Luciferase signal intensity
was also quantified 55 days after injection for siLAMR-transfected cells. siGLO, fluorescently labeled nontargeting siRNA control; siLAMR, siRNA pool
targeting human LAMR; siRNA, small interferin RNA.

receiving siLAMR-transfected tumor cells remained healthy for advanced ovarian cancer animal model.36 Severe combine immu-
the duration of the experiment and after tumor cells regressed nodeficient (SCID) mice were injected intraperitoneally (i.p.) with
(by days 17 or so) did not display luciferase signal by IVIS or any the ES-2 ovarian carcinoma cell and allowed to develop tumors
symptoms associated with recurrence of tumor growth. This dem- in the peritoneal cavity. SCID mice with ES-2-derived tumors
onstrates that LAMR expression plays a major role in the growth and mice without tumors were then treated with a pseudotype
and survival of HT1080 tumors. These results also show, targeting Sindbis/Lenti vector that expresses the firefly luciferase gene. As
and inhibiting LAMR expression in vivo is may be a promising Figure 6b (left) shows, only mice injected with ES-2 cells and have
strategy for treating cancer. tumors show a luciferase signal during mouse whole-body imag-
ing. Further, organs from mice with ES-2 tumors were harvested
Targeting LAMR with Sindbis/Lenti pseudotype from the peritoneal cavity and show that ES-2 tumors and pseudo-
vectors inhibits tumor growth in vivo type Sindbis/Lenti vector localize primarily to the intestine. These
As a proof of principle for altering LAMR expression to treat findings are consistent with our previous results using this model
tumors, we explored the possibility of reducing lamr expression in with Sindbis viral vectors. Our pseudotype vector therefore has
tumor cells in vivo in a targeted manner. We utilized our previous the same tumor-targeting capabilities as Sindbis viral vectors and
experience with Sindbis viral vectors to develop a novel pseudo- can be used for cancer gene therapeutics.
type Sindbis/Lenti vector combining the core proteins of lentivi- We next generated two pseudotype vectors that carry an
rus and the structural proteins of Sindbis virus (Figure 6a). To shRNA cassette, one specific for lamr and another, as a control,
test tumor-targeting capabilities, we used a previously developed specific for LacZ. In vitro transduction of ES-2 ovarian carcinoma

Molecular Therapy vol. 18 no. 1 jan. 2010 69


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

cells with the pseudotype vector specific for lamr reduces LAMR Sindbis/Lenti pseudotype vector to achieve this, SCID mice with
protein expression by ~50% compared to the control vector that tumors derived from ES-2 cells expressing the firefly luciferase
targets LacZ (Figure 6c). Western blots are indicative of at least gene were treated with either the control pseudotype vector or
three independent transductions. To test both the efficacy of reduc- pseudotype vector that reduces LAMR expression. Tumor growth
ing LAMR expression for gene therapy as well as the ability of our was measured by luciferase signal intensity 1 day after injection of

a pLP/E321 PCMV β-Globin intron Sindbis E321 β-Globin pA


b Tumor free Tumor bearing

Sindbis/Lenti
packaging pLP1 PCMV β-Globin intron Gag/Pol RRE β-Globin pA
plasmids

pLP2 PRSV Rev β-Globin pA

PRSV/5’LTR ψ RRE PSV40 EM7 Blasti ∆U3/3’LTR SV40 pA

Expression cassettes
pLenti6/Fluc PUBC Firefly luciferase gene

Lentiviral
expression pLenti6/shLacZ PU6 LacZ shRNA Pol III term
plasmids

pLenti6/shLAMR PU6 LAMR shRNA Pol III term

c LacZ shLAMR
LAMR

β-Actin

HT1080 shLAMR
LAMR protein expression

1.0 LacZ
shLAMR
Fold change

**
0.5

0.0
d Sindbis/Lenti-
shLacZ shLAMR
Day 4 growth percentage
300
Day 1

200
Growth %

60,000 Luciferase whole-body counts


75,000,000

50,000
100 * shLacZ

shLAMR
40,000
50,000,000
Photon counts

0
shLacZ shLAMR
30,000 Treatment
Day 4

20,000 Day 11 growth percentage


3,000 25,000,000

10,000

2,000
Growth %

Counts 0
0 2 4 6 8 10 12
Color Bar
Min = 1,000
* Day
1,000
Max = 60,000
Day 11

0
shLacZ shLAMR
Treatment

70 www.moleculartherapy.org vol. 18 no. 1 jan. 2010


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

ES-2 tumor cells and before pseudotype vector treatment (day 1) regulated in order to induce growth arrest, as can seen by the fact
as well as 3 and 10 days after treatment (day 4 and day 11 after ES-2 that p21 protein levels actually increase while translation as a
tumor-cell injection, respectively) (Figure 6d, left). Luciferase whole is inhibited. It may be that LAMR expression is important
­signal intensity after treatment was compared to signal intensity for translation of a majority of proteins but not all. Further, LAMR
pretreatment to determine tumor growth percentage for day 4 and expression may promote the translation of proteins important for
day 11 (Figure 6d, middle, top, and bottom, respectively). Mice cell-cycle progression, while a reduction in LAMR expression may
receiving treatment with pseudotype vector that targets lamr show lead to translation of proteins that induce growth arrest, such as
~60 and 50% reduced tumor growth compared to control pseudo- p21. Interestingly, it was shown that LAMR/p40 has a higher affin-
type vector on days 4 and 11, respectively. Luciferase whole-body ity for polysomes in plant tissues during times of active growth.37
counts from all days imaged were used to generate tumor growth Therefore, LAMR may only be essential for translation of proteins
curves (Figure 6d, right). The finding that Sindbis/Lenti pseudo- that are required for proliferation.
type vector that reduces LAMR expression also reduces tumor- It is also important to keep in mind that we are working with
cell growth is similar to our previously described results with a transient system, and therefore see slight variations with each
the HT1080 siLAMR–transfected tumor model. Thus, utilizing a transfection. Although we find that siLAMR-transfected cells are
novel vector and approach to cancer gene therapy, we demonstrate always growth arrested, the duration of growth arrest is variable.
that altering LAMR expression can be used for cancer therapy. Likewise, we notice that there are times when siLAMR transfec-
tion has a dramatic effect on cell morphology in vitro (data not
Discussion shown), but this is not always the case. Because of this, the kinetics
Extensive efforts have been made to determine the role of LAMR and specifics of our transient siLAMR system are currently under
in tumor development. Many studies, such as Zuber et al., have further investigation.
focused on extracellular functions including migration and inva- Despite these questions in vitro, the effect of siLAMR in vivo
sion. Experiments performed by our lab have similarly observed is much more profound, indicating that the ribosomal functions
that LAMR is important for both of these processes (data not of LAMR may be more critical for tumor-cell survival in a more
shown). However, our finding that siLAMR-transfected cells have physiological environment. The ability of LAMR to regulate transla-
a limited capacity to proliferate prompted further investigation of tion, proliferation, and survival as a ribosomal protein may there-
LAMR’s potential role in tumor-cell growth. LAMR expression is fore be as important, if not more, than its extracellular functions
essential for cell growth and viability in yeast, and was shown to be in tumor development. This has been suggested by several studies.
critical for ribosome maturation and assembly. Our results have For example, the levels of LAMR upregulation in cervical tumors
found concordance with the yeast studies and clearly show LAMR resulting from papiloma virus infection ­correlates with increased
functions in mammalian cells as a ribosomal protein crucial for cell proliferation but not invasion.38 Also, while LAMR serves as
protein translation. the cellular receptor for the green tea polyphenol (−)-epigallocat-
Protein translation is important for G1 to S phase cell-cycle echin-3-gallate,39 another ribosomal protein, eukaryotic translation
progression. The fact that a reduction in LAMR expression leads elongation factor 1A, is also required for (−)-epigallocatechin-3-
to translation inhibition and G1 cell-cycle arrest indicates LAMR’s ­gallate-induced tumor cell death.40 In addition, (−)-epigallocate-
ribosomal functions can regulate cell proliferation. However, we do chin-3-gallate specifically targets multiple myeloma cell in vivo via
observe that siLAMR-transfected cells start to recover from growth LAMR to induce apoptosis and growth arrest in the G1 phase of the
arrest at a time in which translation is still inhibited, suggesting cell cycle.41 Therefore, the ribosomal functions of LAMR may also be
that perhaps cell proliferation may be regulated by a nonribosomal important for (−)-epigallocatechin-3-gallate-induced effects. These
function of LAMR. It should be noted that the level of translation and our observations add LAMR to a growing list of ribosomal pro-
inhibition appears to be less severe 7 days after ­siLAMR transfec- teins, in which overexpression promotes cell growth and subsequent
tion than at earlier time points when cells are growth arrested. It is loss of LAMR expression (or inhibition) results in cell death.42
possible that the first proteins to be ­translated as cells recover are In this work, our goals were to determine what role LAMR plays
those important for cell-cycle progression. In this case, cells would in translation and tumor viability/development as well as to test its
start to proliferate even as other protein expression is still inhib- therapeutic potential as a target for cancer gene therapy. Recently, it
ited. Conversely, we observe that protein expression is differentially was shown that LAMR expression can be reduced in vivo through

Figure 6 Targeting laminin receptor (LAMR) with Sindbis/Lenti pseudotype vectors inhibits ES-2 tumor growth in vivo. (a) Schematic diagram
of pseudotype vector construct. Notably, vesicular stomatitis virus-G structural proteins have been replaced by Sindbis E1, E2, and E3 structural
proteins. (b) Severe combine immunodeficient (SCID) mice without tumors (tumor free) and SCID mice with peritoneal tumors derived from ES-2
ovarian carcinoma cells (tumor bearing) were treated with a pseudotype Sindbis/Lenti-FLUC vector. In vivo imaging system (IVIS) was used to detect
luciferase expression (pseudotype vector infection) by whole-body imaging (left). Peritoneal organs were extracted from SCID mice with ES-2 tumors
treated with pseudotype Sindbis/Lenti-FLUC and imaged with IVIS to show tumor infection localization (K, kidney; L, liver; S, spleen; I, intestine)
(right). (c) ES-2 ovarian carcinoma cells were transduced in vitro with a Sindbis/Lenti pseudotype vector that carries an shLAMR cassette against lamr
or LacZ. Western blot analysis was used to check expression levels of LAMR and β-actin. Western blots are indicative of at least three separate infections
(top). Protein expression levels were quantified and statistically analyzed (bottom). (d) SCID mice with peritoneal tumors derived from ES-2 FLUC cells
were treated either with a pseudotype vector that targets lamr or a control pseudotype vector that targets LacZ for up to 10 days. Luciferase signal
intensity was imaged by IVIS before (day 1) and after (day 4, day 11) treatment with pseudotype vectors (left). Percentage of growth was quantified
for tumors treated with each pseudotype vector on day 4 and 11 (middle). Growth percentage is the amount of tumor growth compared to day 1.
Luciferase body counts from day 1, 4, and 11 were graphed to generate tumor growth curves (right) (*P < 0.05. **P < 0.001. ***P < 0.0001.).

Molecular Therapy vol. 18 no. 1 jan. 2010 71


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

the use of lentiviral vectors expressing siRNA-­targeting LAMR.43 Materials and Methods
These vectors were used for treatment and prevention of prion dis- Protein analysis. For western blots, cells were harvested in Mammalian
orders. To achieve specificity for tumor targeting, we developed a Protein Extraction Reagent (Pierce, Rockford, IL). A 20 μg of total protein
novel method for targeting LAMR in vivo that utilizes both lenti- was used for sodium dodecyl sulfate–polyacrylamide gel electrophoresis
and transferred to polyvinylidene fluoride membrane. For LAMR protein
viral vector shRNA expression as well as the tumor-targeting capa-
expression, we initially used the goat polyclonal antibody F-18 purchased
bilities of Sindbis viral vectors. We show that LAMR expression from Santa Cruz biotechnology (Santa Cruz, CA) (Figure 1). We later
levels can successfully be diminished through use of our Sindbis/ switched to the rabbit polyclonal antibody H-141 (Santa Cruz) because of
Lenti pseudotype vector and exposure of tumors to this vector its stronger signal. Both recognize epitopes on the C-terminus of LAMR
in vivo caused a reduction of tumor growth as ­predicted. Other and predominantly detect 37-kd LAMR. The Survivin (A-19) antibody
groups have used Sindbis/Lenti pseudotype vectors for different was also purchased from Santa Cruz. The antibody for p21 is from BD
purposes,44–46 some of which were based on our earlier Sindbis- Biosciences (San Jose, CA), and β-actin (AC15) from Sigma-Aldrich
ligand-assisted targeting technology,47 but our studies are the first (St Louis, MO). All other cell-cycle antibodies were gifts from Dr Pagano
at the NYU School of Medicine. Western blots were quantified using The
to demonstrate the use of a Sindbis/Lenti pseudotype vector for
NIH Image 1.63 program (Bethesda, MD).
tumor-targeted knock down of gene expression in animal ­models. To study translation, cells were metabolically labeled with 35S-
By using unmodified Sindbis structural proteins, our pseudotype methionine (20 μCi/ml) (PerkinElmer, Waltham, MA) in Dulbecco’s
vector maintains the natural tumor-targeting capabilities of Sindbis modified Eagle’s medium lacking methionine (MP Biomedicals, Santa
virus through its receptor, LAMR. Thus our Sindbis/Lenti pseudo- Ana, CA) supplemented with 0.1% fetal calf serum for 2 hours at 37 °C
type vector can both target and inhibit tumor growth via binding/ (pulse). Normal growth medium was then added and cells were incubated
downregulating LAMR. for an additional 90 minutes at 37 °C (chase). For trypsin treatement, cells
Targeting LAMR in this study both affirms LAMR’s impor- were incubated with 1× trypsin EDTA from cellgro Mediatech (Manassas,
VA), containing 0.05% trypsin and 0.53 mmol/l EDTA, for ~5 minutes to
tance for cancer therapy and establishes our pseudotype vector as
allow cells to detach. Cells were then reseeded for labeling the next day.
a novel effective therapeutic agent. This represents a potentially
Cells were harvested in Mammalian Protein Extraction Reagent and 20 μg
highly efficacious new therapeutic approach to cancer treatment, of protein was loaded onto sodium dodecyl sulfate–polyacrylamide gel
which can utilize similar pseudotype vectors and approaches to electrophoresis for analysis.
knock down other genes in vivo in a highly targeted manner.
Currently, Sindbis/Lenti-mediated reduction of LAMR is not Gene analysis. Total RNA was extracted from cells using the RNA easy
presently as effective as siRNA. We consistently observe ~50% mini kit (Qiagen, Valencia, CA) and its integrity was analyzed at the NYU
genomics core facility. RNA (0.5 μg) was used for reverse transcription
reduction in LAMR expression with our pseudotype vector
either with Reaction Ready first strand cDNA synthesis kit (SuperArray) or
in vitro, whereas the siLAMR pool achieves ~90% reduction in iScript complementary DNA synthesis kit (BioRad, Hercules, CA). Human
LAMR expression. This less potent reduction in LAMR expres- LAMR and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) prim-
sion is most likely due to the fact that the siRNA pool we use for ers were designed and used as previously described.36 Human GAPDH was
knocking down LAMR consists of four different oligonucleotide chosen as the housekeeping gene for comparative analysis. The fold change
sequences whereas our Sindbis/Lenti vector carries only one. in LAMR relative to the GAPDH endogenous control was determined by:
Generating pseudotype vectors that carry more than one shRNA fold change = 2(–Ct), where CT = CT(LAMR) − CT(GAPDH) and (CT) = CT(ES-2/FLUC/
sequence may help increase its knock-down efficiency. In addition, LRP)
− CT(ES-2/FLUC). CT is the threshold cycle determined for fluorescence data
collection. GAPDH and this formula were also used for analysis of cell
we have not yet been able to produce the Sindbis/Lenti pseudotype
cycle–related genes. The cell-cycle RT2Profiler PCR array and primers for
vectors at as high a titer as we can achieve with Sindbis ­vectors validation of cyclin A2, cyclin B1, CDK2, E2F1, p21, Survivin, as well as an
resulting in a transduction efficiencies that are not as high in vivo, additional GAPDH primer set were purchased from SuperArray. Primers
thus some tumor cells are not transduced and hence remain via- for CDK1 were obtained online from Primer Bank.49 Quantitative—real-
ble. Further efforts to optimize the vectors described here will time PCR was performed with 0.5 μl of complementary DNA using a
develop more potent vectors targeting LAMR as well as delivery of BioRad iCycler.
shRNA in vivo for additional therapeutic purposes. Finally, alter-
native strategies that target LAMR in a ribosomal context may Cell culture. HT1080, 293, HeLa, and HepG2, and ES-2 cells were obtained
from the ATCC (Manassas, VA). ES-2/FLUC cells were derived using a
also be employed and our findings are supportive of the notion
pIRES2-Luc/EGFP plasmid as previously described.36 To ­generate HT1080
that these efforts might ultimately be very advantageous. To this FLUC cells, the FLUC gene was amplified by PCR from the pIRES2-
end, our lab has recently solved the crystal structure of an LAMR Luc/EGFP plasmid and cloned into the TOPO pcDNA3.1v5/his vector
construct consisting of residues 1–220 (ref. 48) and are currently (Invitrogen, Carlsbad, CA) to create a pcDNA3.1/FLUCv5/his plasmid.
using this structure as well as the system established by our cur- HT1080 cells were transfected with this plasmid and clones were selected
rent results in pursuing the design of small molecules to inhibit with neomycin. The predesigned siGENOME SMART pool ­targeting LAMR,
LAMR ­functions. It is likely that a combination of both vector- siGLO RISC-free siRNA, and DharmaFECT transfection reagents were
and drug-based modalities will optimize treatment. purchased from Dharmacon (Lafayette, CO). Individual oligo sequences
and the region of LAMR they target are provided (Supplementary Figure
In conclusion, the studies reported here point to several mech-
S1). A final concentration of 100 nmol/l of siRNA was used for each
anisms by which LAMR can contribute to tumor development; transfection. To determine siGLO transfection efficiency, nontransfected
further underscoring its importance as a target for cancer ther- and siGLO-transfected cells were analyzed 1 day after transfection on a
apy. In addition, we provide a novel mechanism for gene therapy FACSCaliber machine. The Flowjo 8.2 program (Tree Star, Ashland, OR)
in vivo, which can be used against LAMR and potentially other was used to quantify the percentage of siGLO-transfected cells positive in
genes as well. the FL2-H channel, compared to nontransfected cells.

72 www.moleculartherapy.org vol. 18 no. 1 jan. 2010


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

Cells were imaged with a Nikon Eclipse TE200-E microscope Animals. For Sindbis/Lenti pseudotype–targeting experiments, ES-2
(Tokyo, Japan) using the NIS-Elements BR-2.30 program (Nikon). Cell human ovarian carcinoma cells (2 million) were inoculated i.p. into female
proliferation was measured using the CellTiter-Glo Cell Viability assay SCID mice (4–8 weeks old, Taconic Farms, Hudson, NY). At 7, 8, and 9
from (Promega, Madison, WI) and luminescence was measured on a days after injection of ES-2 cells, mice were treated with Sindbis/­Lenti-
GLOMAX 20/20 luminometer (Promega). For cell-cycle profile analysis, FLUC vector (1 cm3, 106 plaque-forming units) by i.p. injection. At 10 days
cells were permeabilized overnight in 100% ethanol and then stained with after ES-2 injection, and 3 days of pseudotype vector treatment, mice were
final concentrations of 50 μg/ml of propidium iodide and 100 μg/ml of subjected to IVIS imaging. For Sindbis/Lenti shLAMR experiments, ES2-
RNAse A (Sigma, St Louis, MO). Cells were then submitted to the NYU FLUC cells (3 million) were inoculated i.p. into female SCID mice (Taconic
Cancer institute core and analyzed on a FACSCaliber machine. Farms). One day after inoculation, first IVIS images were taken to establish
baseline tumor load. Mice were then treated daily i.p. with 0.5 ml of Sindbis/
Ribosomal analysis. Ribosomes and polyribosomes were isolated and Lenti-shLacZ or Sindbis/Lenti-shLAMR (~106 plaque-forming units) for
analyzed as described by Rouquette et al.50 Briefly, siLAMR- and siGLO- three consecutive days. After treatment, 4 days after ES-2 inoculation, mice
­transfected cells were treated with 100 μg/ml of cyclohexamide for 10 were subjected to a second IVIS imaging. Mice were then treated daily i.p.
­minutes at 37 °C. An equal number of cells were pelleted and lysed in lysis with 0.5 ml of Sindbis/Lenti-shLacZ or Sindbis/Lenti-shLAMR (~106) for
buffer with a Dounce homogenizer. Cell lysates were spun at 14,000 r.p.m. four consecutive days, 7–10 days after ES-2 injection. At 11 days after ES-2
for 10 minutes to remove cellular debris and then layered onto 10–50% injection mice were subjected to a third IVIS imaging.
linear sucrose gradients (prepared ~16 hours before use). Gradients were
spun for 105 minutes at 40,000g in a SW41TI rotor (Beckman, Brea, CA). In vivo bioluminescence detection with the IVIS. A cryogenically cooled
A total of 24 (500-μl) fractions were collected from the top of each gradient IVIS Spectrum Imaging System (Caliper LifeScience, Alameda, CA) with
and optical density was measured at A260 nm. In addition, 32 μl of fractions Living Image 3.0 acquisition and analysis software (version 2.11; Xenogen,
1–12 were added to 8 μl of 5× reducing buffer and used for western blot Alameda, CA) was used to detect the bioluminescence signals in mice.
analysis. Antibodies for ribosomal proteins L7a and S6 were purchased Each mouse was injected i.p. with 0.3 ml of 15 mg/ml beetle luciferin
from Cell Signaling (Danvers, MA). (potassium salt; Promega) in phosphate-buffered saline. After 5 minutes,
mice were anesthetized with isofluoran mixed oxygen (2%). The imaging
Viral vector construction and production. To produce Sindbis/Lenti system first took a photographic image in the chamber under dim illumi-
pseudotype vectors, three packaging plasmids (pLP1, pLP2, and pLP/ nation, followed by luminescent image acquisition. Overlay of the pseudo-
E321), which provide structural proteins, and the expression plasmid color images represents the spatial distribution of photon counts produced
(pLenti6), encoding a modified lentiviral RNA genome, were used. Sindbis by active luciferase. An integration time of 1 minute at medium binning
viral envelope proteins (E3, E2, and E1) are provided by the pLP/E321 was used for luminescent image acquisition for all animal tumor models.
plasmid. To generate the pLP/E321 plasmid, the E321 gene fragment was Living Image 3.0 software was used to integrate the total bioluminescence
first amplified by PCR using pDHBB plasmid as a template, which contains signals (in terms of photon counts) obtained from animals.
the full length of Sindbis structural proteins. The PCR product was first
cloned into pcDNA3.1 plasmid (Invitrogen) for sequencing validation and Statistical analysis. Data were analyzed using a standard Student’s t-test
was later used to replace the vesicular stomatitis virus-G fragment (from using GraphPad Prism, version 3.0a for Macintosh (GraphPad Software,
StuI to PlmI site) in the pLP/VSVG plasmid. San Diego, CA). All P values presented in this study are two-tailed. In
pLenti6 carrying an ubiquitin C promoter (PUBC) was used to drive our analysis, results for experimental groups (siLAMR or shLAMR) were
Pol II-dependent expression of the firefly luciferase gene (pLenti6/FLUC). considered statistically significant relative to the control group (siGLO or
A U6 shRNA promoter was used for shRNA expression (pLenti6/shLacZ shLacZ) if the P value <0.05.
or pLenti6/shLAMR). The DNA oligos containing the corresponding
shRNA sequences against LacZ or LAMR genes were first cloned into the Supplementary Material
Figure S1. siLAMR targeting sequences within the lamr coding region.
pENTR/U6 plasmid and sequenced before recombination with pLenti6/
Block-iT-DEST plasmid according to manufacturer’s instruction. Acknowledgments
Oligos used are as follows: US Public Health grants CA100687 from the National Cancer Institute,
shLacZ top: 5′-CACCGCTACACAAATCAGCGATTTCGAAAAAT National Institutes of Health, and Department of Health and Human
CGCTGATTTGTGTAG-3′. Services supported this study. Funding was also provided by a gift
shLacZ bottom: 5′-AAAACTACACAAATCAGCGATTTTTCGAAA from the Litwin Foundation and a Research and License agreement be-
TCGCTGATTTGTGTAGC-3′. tween NYU and CynVec. Some of the authors have competing inter-
shLAMR top: 5′-CACCGCAACAAGGGAGCTCACTCACGAATG ests. Specifically, the contents of this study may be utilized for a patent.
AGTGAGCTCCCTTGTTG-3′. According to the rules and regulations of New York University School of
shLAMR bottom: 5′-AAAACAACAAGGGAGCTCACTCATTCGT Medicine, if this patent is licensed by a third party, some of the authors
GAGTGAGCTCCCTTGTTGC-3′. may receive benefits in the form of royalties or equity participation.
Packaging plasmids (pLP1, pLP2, and pLP/E321) and a pLent6 plasmid
(pLenti6/FLUC, pLenti6/shLacZ, or pLenti6/shLAMR) were co-transfected References
1. Gauczynski, S, Peyrin, JM, Haïk, S, Leucht, C, Hundt, C, Rieger, R et al. (2001).
into 293T cells using the calcium phosphate method. Pseudotype vector The 37-kDa/67-kDa laminin receptor acts as the cell-surface receptor for the cellular
particles were collected in supernatant and pelleted using ultracentrifugation prion protein. EMBO J 20: 5863–5875.
2. Kim, KJ, Chung, JW and Kim, KS (2005). 67-kDa laminin receptor promotes
at 25,000 r.p.m. for 2 hours with an SW-28 rotor. The vector pellets were internalization of cytotoxic necrotizing factor 1-expressing Escherichia coli K1 into
diluted into ×1/50 volume before centrifugation using Dulbecco’s modified human brain microvascular endothelial cells. J Biol Chem 280: 1360–1368.
3. Wang, KS, Kuhn, RJ, Strauss, EG, Ou, S and Strauss, JH (1992). High-affinity laminin
Eagle’s medium. receptor is a receptor for Sindbis virus in mammalian cells. J Virol 66: 4992–5001.
The capability of Sindbis/Lenti-shLAMR to suppress LAMR expression 4. Thepparit, C and Smith, DR (2004). Serotype-specific entry of dengue virus into liver
was tested in culture using ES2/FLUC cells. ES2/FLUC cells were seeded onto cells: identification of the 37-kilodalton/67-kilodalton high-affinity laminin receptor
as a dengue virus serotype 1 receptor. J Virol 78: 12647–12656.
6-well plates the day before transduction. The next day, 100 μl virus plus 400 μl 5. Ludwig, GV, Kondig, JP and Smith, JF (1996). A putative receptor for Venezuelan
of medium was added to the wells and incubated for 24 hours. The vector was equine encephalitis virus from mosquito cells. J Virol 70: 5592–5599.
6. Akache, B, Grimm, D, Pandey, K, Yant, SR, Xu, H and Kay, MA (2006). The 37/67-
then removed and replaced with fresh medium and incubated for 48 hours. kilodalton laminin receptor is a receptor for adeno-associated virus serotypes 8, 2,
Cells were lysed and LAMR expression determined by western blot. 3, and 9. J Virol 80: 9831–9836.

Molecular Therapy vol. 18 no. 1 jan. 2010 73


© The American Society of Gene & Cell Therapy
LAMR as a Target for Novel Cancer Gene Therapy

7. van den Brule, FA, Buicu, C, Berchuck, A, Bast, RC, Deprez, M, Liu, FT et al. (1996). 29. Kaneda, Y, Kinoshita, K, Sato, M, Saeki, Y, Yamada, R, Wataya-Kaneda, M et al. (1998).
Expression of the 67-kD laminin receptor, galectin-1, and galectin-3 in advanced The induction of apoptosis in HeLa cells by the loss of LBP-p40. Cell Death Differ 5:
human uterine adenocarcinoma. Hum Pathol 27: 1185–1191. 20–28.
8. Taraboletti, G, Belotti, D, Giavazzi, R, Sobel, ME and Castronovo, V (1993). 30. Susantad, T and Smith, DR (2008). siRNA-mediated silencing of the 37/67-kDa
Enhancement of metastatic potential of murine and human melanoma cells by high affinity laminin receptor in Hep3B cells induces apoptosis. Cell Mol Biol Lett 13:
laminin receptor peptide G: attachment of cancer cells to subendothelial matrix as a 452–464.
pathway for hematogenous metastasis. J Natl Cancer Inst 85: 235–240. 31. Kinoshita, K, Kaneda, Y, Sato, M, Saeki, Y, Wataya-Kaneda, M and Hoffmann, A
9. Sanjuán, X, Fernández, PL, Miquel, R, Muñoz, J, Castronovo, V, Ménard S et al. (1998). LBP-p40 binds DNA tightly through associations with histones H2A, H2B,
(1996). Overexpression of the 67-kD laminin receptor correlates with tumour and H4. Biochem Biophys Res Commun 253: 277–282.
progression in human colorectal carcinoma. J Pathol 179: 376–380. 32. Auth, D and Brawerman, G (1992). A 33-kDa polypeptide with homology to the
10. Montuori, N, Selleri, C, Risitano, AM, Raiola, AM, Ragno, P, Del Vecchio, L et al. laminin receptor: component of translation machinery. Proc Natl Acad Sci USA 89:
(1999). Expression of the 67-kDa laminin receptor in acute myeloid leukemia 4368–4372.
cells mediates adhesion to laminin and is frequently associated with monocytic 33. Ludewigs, H, Zuber, C, Vana, K, Nikles, D, Zerr, I and Weiss, S (2007). Therapeutic
differentiation. Clin Cancer Res 5: 1465–1472. approaches for prion disorders. Expert Rev Anti Infect Ther 5: 613–630.
11. Martignone, S, Ménard, S, Bufalino, R, Cascinelli, N, Pellegrini, R, Tagliabue, E et al. 34. Zuber, C, Knackmuss, S, Zemora, G, Reusch, U, Vlasova, E, Diehl, D et al. (2008).
(1993). Prognostic significance of the 67-kilodalton laminin receptor expression Invasion of tumorigenic HT1080 cells is impeded by blocking or downregulating the
in human breast carcinomas. J Natl Cancer Inst 85: 398–402. 37-kDa/67-kDa laminin receptor. J Mol Biol 378: 530–539.
12. Fontanini, G, Vignati, S, Chiné, S, Lucchi, M, Mussi, A, Angeletti, CA et al. (1997). 35. Narumi, K, Inoue, A, Tanaka, M, Isemura, M, Shimo-Oka, T, Abe, T et al. (1999).
67-Kilodalton laminin receptor expression correlates with worse prognostic indicators Inhibition of experimental metastasis of human fibrosarcoma cells by anti-
in non-small cell lung carcinomas. Clin Cancer Res 3: 227–231. recombinant 37-kDa laminin binding protein antibody. Jpn J Cancer Res 90: 425–431.
13. Wewer, UM, Taraboletti, G, Sobel, ME, Albrechtsen, R and Liotta, LA (1987). Role of 36. Tseng, JC, Hurtado, A, Yee, H, Levin, B, Boivin, C, Benet, M et al. (2004). Using sindbis
laminin receptor in tumor cell migration. Cancer Res 47: 5691–5698. viral vectors for specific detection and suppression of advanced ovarian cancer in
14. Bernard, A, Gao-Li, J, Franco, CA, Bouceba, T, Huet, A and Li, Z (2009). Laminin animal models. Cancer Res 64: 6684–6692.
receptor involvement in the anti-angiogenic activity of pigment epithelium-derived 37. Garcia-Hernandez, M, Davies, E, Baskin, TI and Staswick, PE (1996). Association of
factor. J Biol Chem 284: 10480–10490. Plant p40 Protein with Ribosomes Is Enhanced When Polyribosomes Form during
15. Berno, V, Porrini, D, Castiglioni, F, Campiglio, M, Casalini, P, Pupa, SM et al. (2005). Periods of Active Tissue Growth. Plant Physiol 111: 559–568.
The 67 kDa laminin receptor increases tumor aggressiveness by remodeling laminin-1. 38. Demeter, LM, Stoler, MH, Sobel, ME, Broker, TR and Chow, LT (1992). Expression
Endocr Relat Cancer 12: 393–406. of high-affinity laminin receptor mRNA correlates with cell proliferation rather than
16. Givant-Horwitz, V, Davidson, B and Reich, R (2004). Laminin-induced signaling invasion in human papillomavirus-associated cervical neoplasms. Cancer Res 52:
in tumor cells: the role of the M(r) 67,000 laminin receptor. Cancer Res 64: 1561–1567.
3572–3579. 39. Tachibana, H, Koga, K, Fujimura, Y and Yamada, K (2004). A receptor for green tea
17. Ménard, S, Tagliabue, E and Colnaghi, MI (1998). The 67 kDa laminin receptor as a polyphenol EGCG. Nat Struct Mol Biol 11: 380–381.
prognostic factor in human cancer. Breast Cancer Res Treat 52: 137–145. 40. Umeda, D, Yano, S, Yamada, K and Tachibana, H (2008). Green tea polyphenol
18. Rao, NC, Barsky, SH, Terranova, VP and Liotta, LA (1983). Isolation of a tumor cell epigallocatechin-3-gallate signaling pathway through 67-kDa laminin receptor.
laminin receptor. Biochem Biophys Res Commun 111: 804–808. J Biol Chem 283: 3050–3058.
19. Malinoff, HL and Wicha, MS (1983). Isolation of a cell surface receptor protein for 41. Shammas, MA, Neri, P, Koley, H, Batchu, RB, Bertheau, RC, Munshi, V et al.
laminin from murine fibrosarcoma cells. J Cell Biol 96: 1475–1479. (2006). Specific killing of multiple myeloma cells by (-)-epigallocatechin-3-gallate
20. Lesot, H, Kühl, U and Mark, KV (1983). Isolation of a laminin-binding protein from extracted from green tea: biologic activity and therapeutic implications. Blood
muscle cell membranes. EMBO J 2: 861–865. 108: 2804–2810.
21. Rao, CN, Castronovo, V, Schmitt, MC, Wewer, UM, Claysmith, AP, Liotta, LA et al. 42. Naora, H (1999). Involvement of ribosomal proteins in regulating cell growth and
(1989). Evidence for a precursor of the high-affinity metastasis-associated murine apoptosis: translational modulation or recruitment for extraribosomal activity?
laminin receptor. Biochemistry 28: 7476–7486. Immunol Cell Biol 77: 197–205.
22. Landowski, TH, Dratz, EA and Starkey, JR (1995). Studies of the structure of the 43. Pflanz, H, Vana, K, Mitteregger, G, Pace, C, Messow, D, Sedlaczek, C et al. (2009).
metastasis-associated 67 kDa laminin binding protein: fatty acid acylation and Microinjection of lentiviral vectors expressing small interfering RNAs directed against
evidence supporting dimerization of the 32 kDa gene product to form the mature laminin receptor precursor mRNA prolongs the pre-clinical phase in scrapie-infected
protein. Biochemistry 34: 11276–11287. mice. J Gen Virol 90(Pt 1): 269–274.
23. Butò, S, Tagliabue, E, Ardini, E, Magnifico, A, Ghirelli, C, van den Brûle, F et al. 44. Yang, L, Yang, H, Rideout, K, Cho, T, Joo, KI, Ziegler, L et al. (2008). Engineered
(1998). Formation of the 67-kDa laminin receptor by acylation of the precursor. lentivector targeting of dendritic cells for in vivo immunization. Nat Biotechnol 26:
J Cell Biochem 69: 244–251. 326–334.
24. Hundt, C, Peyrin, JM, Haïk, S, Gauczynski, S, Leucht, C, Rieger, R et al. (2001). 45. Morizono, K, Xie, Y, Ringpis, GE, Johnson, M, Nassanian, H, Lee, B et al. (2005).
Identification of interaction domains of the prion protein with its 37-kDa/67-kDa Lentiviral vector retargeting to P-glycoprotein on metastatic melanoma through
laminin receptor. EMBO J 20: 5876–5886. intravenous injection. Nat Med 11: 346–352.
25. Ardini, E, Pesole, G, Tagliabue, E, Magnifico, A, Castronovo, V, Sobel, ME et al. (1998). 46. Aires da Silva, F, Costa, MJ, Corte-Real, S and Goncalves, J (2005). Cell type-specific
The 67-kDa laminin receptor originated from a ribosomal protein that acquired a dual targeting with sindbis pseudotyped lentiviral vectors displaying anti-CCR5 single-chain
function during evolution. Mol Biol Evol 15: 1017–1025. antibodies. Hum Gene Ther 16: 223–234.
26. Demianova, M, Formosa, TG and Ellis, SR (1996). Yeast proteins related to the p40/ 47. Ohno, K, Sawai, K, Iijima, Y, Levin, B and Meruelo, D (1997). Cell-specific targeting
laminin receptor precursor are essential components of the 40 S ribosomal subunit. of Sindbis virus vectors displaying IgG-binding domains of protein A. Nat Biotechnol
J Biol Chem 271: 11383–11391. 15: 763–767.
27. García-Hernández, M, Davies, E and Staswick, PE (1994). Arabidopsis p40 homologue. 48. Jamieson, KV, Wu, J, Hubbard, SR and Meruelo, D (2008). Crystal structure of the
A novel acidic protein associated with the 40 S subunit of ribosomes. J Biol Chem 269: human laminin receptor precursor. J Biol Chem 283: 3002–3005.
20744–20749. 49. Wang, X and Seed, B (2003). A PCR primer bank for quantitative gene expression
28. Ford, CL, Randal-Whitis, L and Ellis, SR (1999). Yeast proteins related to analysis. Nucleic Acids Res 31: e154.
the p40/laminin receptor precursor are required for 20S ribosomal RNA 50. Rouquette, J, Choesmel, V and Gleizes, PE (2005). Nuclear export and cytoplasmic
processing and the maturation of 40S ribosomal subunits. Cancer Res 59: processing of precursors to the 40S ribosomal subunits in mammalian cells.
704–710. EMBO J 24: 2862–2872.

74 www.moleculartherapy.org vol. 18 no. 1 jan. 2010

You might also like