Hamsa 2010

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

European Journal of Pharmacology 649 (2010) 64–73

Contents lists available at ScienceDirect

European Journal of Pharmacology


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / e j p h a r

Molecular and Cellular Pharmacology

Harmine inhibits tumour specific neo-vessel formation by regulating VEGF, MMP,


TIMP and pro-inflammatory mediators both in vivo and in vitro
T.P. Hamsa, Girija Kuttan ⁎
Amala Cancer Research Centre, Amala Nagar, Thrissur, 680 555, Kerala, India

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

Article history: Harmine is a beta-carboline alkaloid present in medicinal plants such as Peganum harmala that have been used as
Received 15 April 2010 folk medicine in anticancer therapy. In this study, we demonstrated the anti-angiogenic activity of harmine using
Received in revised form 21 August 2010 in vivo and in vitro assay systems. In vivo anti-angiogenic activity was studied using B16F-10 melanoma cells
Accepted 9 September 2010
which induced capillary formation in C57BL/6 mice. Intraperitoneal administration of harmine at 10 mg/kg body
Available online 19 September 2010
weight significantly decreased tumour directed capillary formation. A drastic elevation in serum pro-angiogenic
Keywords:
factors such as vascular endothelial growth factor (VEGF), nitric oxide (NO) and pro-inflammatory cytokines in
Harmine angiogenesis induced animals was significantly decreased by harmine treatment. At the same time harmine
Anti-angiogenesis increased anti-tumour factors like interleukin-2 (IL-2) and tissue inhibitor metalloprotease (TIMP). Moreover
VEGF nuclear factor (NF)-κB and other transcription factors like CREB, ATF-2 involved in tumour development and
B16F-10 melanoma angiogenesis were also inhibited by harmine. Various in vitro assays also supported the anti-angiogenic activity of
Pro-inflammatory cytokine harmine. It reduced proliferation, migration and tube formation of human umbilical vein endothelial cells
Transcription factor (HUVEC). Direct treatment of the harmine also inhibited microvessel outgrowth from the rat aortic ring.
Production of other factors by tumour cells which are involved in angiogenesis like cyclooxygenase (COX-2),
inducible nitric oxide synthase (iNOS) and matrix metalloproteases (MMPs) were also decrease by the treatment
with harmine. Our data suggest that harmine may be a strong angiogenic inhibitor with the ability to decrease the
proliferation of vascular endothelial cells and to reduce expression of various pro-angiogenic factors.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction cellular matrix (ECM). Local degradation of the vascular basement


membrane and ECM occur simultaneously by matrix metallopro-
Angiogenesis, the formation of new blood vessels from pre- teases (MMP) released by tip cells paving the way for newly sprouting
existing vasculature, is prevalent both during normal mammalian vessels (Adams and Alitalo, 2007).
development and in certain pathological conditions such as tumour Inflammation has been found as one of the important processes in
growth. Angiogenesis involves a sequence of coordinated events that mediating angiogenesis. For example, many tumour cell types and
is initiated by the expression of angiogenic factors such as vascular tumour-infiltrating macrophages express tumour necrosis factor (TNF-
endothelial growth factor (VEGF). VEGF increases vascular perme- α) and interleukins (IL). These molecules promote and sustain the
ability, which leads to extravasation of plasma proteins and production of the classical angiogenic factors VEGF-A and basic fibroblast
dissociation of pericyte coverage (Dvorak, 2005; Roberts and Palade, growth factor (bFGF) and mediates the ECM remodeling through the
1997). VEGF promotes endothelial proliferation via the activation of induction of metalloproteases, such as MMP-9 (Sethi et al., 2008).
the MAPK, Akt and inducible nitric oxide synthase (iNOS) pathways Inhibiting the angiogenic process or targeting existing tumour vessels
(Lal et al., 2001; Zhu et al., 2000). One of the first events to take place can be used for the treatment of tumours as an alternative or in parallel
during the angiogenic process involves the formation of a vascular with conventional chemotherapy. The therapeutic strategy to develop
sprout. This begins with the conversion of endothelial cells into an anti-angiogenic molecules involve various targets including angiogenic
endothelial tip cell which transforms the cell from a quiescent into an molecules from tumour cells and inflammatory cells such as VEGF, bFGF,
actively migrating state. Endothelial tip cells undergo extensive TNF-α, IL-8, etc. and their respective receptors on endothelial cells,
migration and proliferation under the influence of VEGF and extra endothelial cells itself, MMPs, COXs etc. (Bhat and Singh, 2008).
Endogenous angiogenesis inhibitors such as interferons (IFN), tissue
inhibitor metalloproteases (TIMP), and proteolytic fragments also act as
⁎ Corresponding author. Department of Immunology, Amala Cancer Research Centre, target for anti-angiogenic therapy (Feldman and Libutti, 2000). TIMP
Amala Nagar, Thrissur. 680 555, Kerala, India. Tel.: + 91 487 2307968; fax: + 91 487
2307868.
inhibits the activities of MMP-1, MMP-2 and MMP-9, which leads to
E-mail addresses: amalacanceresearch@gmail.com, girijakuttan@gmail.com reduced ECM remodeling and suppression of endothelial cell migration
(G. Kuttan). and invasion (Gomez et al., 1997a).

0014-2999/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.ejphar.2010.09.010
T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73 65

Beta-carboline alkaloids such as harmine (Fig. 1) are present in 2.4. Chemicals


medicinal plants such as Peganum harmala that have been used as folk
medicine in anticancer therapy (Berrougui et al., 2006). It is known MTT (3-4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bro-
that these alkaloids have a wide spectrum of neuropharmacological, mide), DMEM, and oligonucleotide primer sequences were purchased
psychopharmacological (Abe and Yamada, 2009), anti-tumour, anti- from Sigma, St. Louis, USA. All the other reagents and chemicals used
bacterial and anti-plasmodial effect (Arshad et al., 2008). Therefore, were of analytical reagent grade.
they have been traditionally used in oriental medicine for the
treatment of various diseases including cancers. It was reported that
2.5. Drug administration
harmine showed significant tumour inhibition in mice bearing Lewis
Lung Cancer, sarcoma180 or HepA tumour (Chen et al., 2005), and
Harmine was dissolved in sterile water. A non toxic dose of these
showed broad cytotoxicity spectrum against KB, SaOS-2, A549, U-87-
compounds were administered intraperitoneally (i.p.) at a dosage of
MG, and MCF-7 cells(Ishida et al., 1999) and human lung carcinoma
10 mg/kg body weight of animal and for in vitro experiments harmine
cell lines (Abe and Yamada, 2009). Studies have demonstrated that
was dissolved in dimethyl sulfoxide (DMSO) (0.1%) and resuspended
these alkaloids can also act as scavengers of reactive oxygen species
in culture medium. TNP-470 (from Dr. Ravivarma, NCI, USA) also
(Moura et al., 2007). On the basis of these potential anticancer
dissolved in DMSO (0.1%), resuspended in sterile water and
activities of harmine, in this study we analyzed its effect on tumour
administered intraperitoneally as a reference compound.
specific angiogenesis and its mechanism of action in both in vivo as
well as in vitro models. Various in vitro assays such as endothelial cell
matrix degradation, migration, proliferation and in vivo studies like 2.6. Effect of harmine on tumour specific capillary formation
rat aortic ring assay, tumour directed capillary formation analysis are
used to assess the anti-angiogenic activity of harmine. Various pro and Angiogenesis was induced in three groups of mice (n = 8) by
anti-angiogenic factors in the presence and absence of harmine are injecting B16F-10 melanoma cells (106 cells/animal) intradermally on
also analyzed by this experiment. the shaven ventral skin of each mouse and were treated simultaneously
(i.p.) with 10 mg harmine per kg body weight or 30 mg TNP-470 per kg
body weight for five consecutive days. Control animals received B16F-10
2. Materials and methods cells alone. After 24 h and 9 days of tumour challenge, blood was
collected in a sterile manner from the caudal vein of each mouse. Serum
2.1. Animals was separated and used for the estimation of cytokines using ELISA kits
according to the manufacturers' instructions. After 9 days, the animals
Four to six week old male C57BL/6 mice were purchased from were sacrificed and the skin from the ventral side was dissected out,
National Institute of Nutrition, Hyderabad, India. The animals were fed washed with PBS and the number of capillaries growing towards the site
with normal mouse chow (Sai Feeds, India) and water ad libitum. All of tumour induction from the pre-existing vessels were counted using a
animal experiments were conducted according to the rules and dissection microscope at 20× magnification and compared with the
regulations of Animal Ethics Committee, Govt. of India. Human untreated control animals (Leyon and Kuttan, 2004).
umbilical vein endothelial cell (HUVEC) experiments were conducted
according to the guidelines of institutional ethical committee.
2.7. Effect of harmine on the production of IL-1β, IL-6, TNF-α, GM-CSF,
IL-2, VEGF and TIMP-1 during angiogenesis
2.2. Cells
Blood obtained from the above experiment at two time intervals
HUVECs were isolated from human umbilical cord vein by was centrifuged and serum was separated. This serum was used for
collagenase treatment as described by Jaffe et al. (Jaffe et al., 1973). the estimation of IL-1β, IL-6, TNF-α, GM-CSF, IL-2, VEGF and TIMP-1
The cells were grown in medium 199, supplemented with 20% fetal using ELISA kits according to manufacturer's protocol.
Bovine serum (FBS), 100 units/ml penicillin, 100 μg/ml streptomycin
and 2 ng/ml VEGF and FGF at 37 °C in 5% CO2 atmosphere. B16F-10
2.8. Effect of harmine on serum nitrite levels during angiogenesis
melanoma cells were purchased from the National Centre for Cell
Science, Pune, India and maintained in culture using DMEM with 10%
Serum obtained from the above experiment at two time intervals
FBS and antibiotics.
was also used for the estimation of nitrite level by Griess reaction
(Green et al., 1982). Briefly a 100 μl sample was incubated with an
equal amount of Griess reagent (one part of 0.1% N(1-naphthyl)-
2.3. ELISA kits
diamine dihydrochloride in distilled water and one part 1% sulfanil-
amide in 5% concentrated H3PO4) and incubated for 10 min at room
Highly specific quantitative ‘sand witch’ Elisa kits for mouse IL-1β,
temperature. Absorbance was estimated at 540 nm. The amount of
IL-6, TNF-α, GM-CSF and IL-2 were purchased from Pierce Biotech-
nitrite was calculated from the NaNO2 standard curve.
nology USA and the ELISA kits for VEGF and TIMP-1 were purchased
from R&D Systems, USA.
2.9. Cell viability by MTT assay

The viability of cultured cells was determined by assaying for the


reduction of MTT to formazan (Campling et al., 1991; Cole, 1986).
HUVECs were seeded (5000 cells/well) in titre plate and incubated for
24 h at 37 °C in 5% CO2 atmosphere. Different concentrations of
harmine (1 μg–50 μg/ml) were added and incubated further for 48 h.
Before the 4 h completion of incubation, 20 μl MTT (5 mg/ml) was
added. The percentage of viable cells was determined using an ELISA
Fig. 1. Structure of harmine. plate reader set to record absorbance at 570 nm.
66 T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73

2.10. Effect of harmine on endothelial cell proliferation (3H-thymidine incubated with or without different concentrations of harmine (0.5, 1
incorporation assay) and 2 μg/ml). In a second set of experiment, the aortic rings were
incubated for 24 h in complete medium initially, and then replaced with
HUVECs (5000 cells/well) were plated in a 96-well culture plate and CM from harmine treated (pretreated CM) and non-treated B16F-10
incubated at 37 °C in 5% CO2 atmosphere. After 24 h, various melanoma cells. On day 6, the rings were analyzed for microvessel
concentrations of harmine (0.5, 1 and 2 μg/ml) were added along with outgrowth and photographed.
2 ng/ml VEGF and further incubated for 48 h. 3H-thymidine was added
to each well (1 μCi/well) and incubation was continued for additional 2.15. Quantitation of gene specific mRNA of VEGF in B16F-10 melanoma
18 h. After completing incubation, the plates were centrifuged and the cells
culture supernatant was removed, the cells were washed three times
with PBS and then treated with ice cold percholric acid for 15 min. The Quantikine mRNA is a novel method, which can be used to
resulting precipitate was dissolved in 0.5 N NaOH and was added to the quantitate gene specific mRNA at lower levels. Briefly, B16F-10 cells
scintillation fluid and the radioactivity was counted using a Rack Beta (106 cells) were plated in 30 mm petridish in DMEM with 10% FBS at
liquid scintillation counter. 37 °C in 5% CO2 atmosphere. Cells were then treated with harmine
(2 μg/ml) for 4 h. After incubation, the cells were washed and mRNA
2.11. Effect of harmine on endothelial cell migration/motility preparations were made according to the manufacturer's procedures.
mRNAs were hybridized with gene specific biotin labeled detection
HUVECs were seeded into wells of collagen coated 96-well flat probes and digoxigenin alkaline labeled detection probes in a
bottom plates at a density of 2 × 105 cells/well and incubated for 24 h microplate. Hybridization solution was transferred to a streptavidin
at 37 °C in 5% CO2 atmosphere. A clear area was scraped in the coated microplate and the mRNA probe hybrid was captured. Fol-
monolayer with a narrow tip by applying suction and washed with lowing wash to remove the unbound conjugate, a substrate solution
serum free medium. Different concentrations of harmine (0.5, 1 and was added. An amplifier solution was then added and the developed
2 μg/ml) were added along with 2 ng/ml VEGF and further incubated colour was measured spectrophotometrically at 490 nm.
for 24 h. After incubation the cells were fixed in formalin and stained
with crystal violet and photographed (200×) (Guo et al., 2002). 2.16. Effect of harmine on NF-κB, c-Fos, ATF-2 and CREB-1

2.12. Effect of harmine on endothelial cell invasion 2.16.1. Preparation of nuclear extracts
Nuclear extracts were prepared by the previously published
The invasion assay was carried out in modified Boyden chambers as method (Dignam et al., 1983). B16F-10 cells were treated with
described by Albini et al. (1987). The lower compartment of the harmine (2 μg/ml) for 2 h at 37 °C in 5% CO2 atmosphere in serum free
chamber was filled with serum free medium 199 and a polycarbonate medium. The cells were washed twice with PBS and incubated further
filter membrane coated with 25 μg Type I collagen was placed above with TNF-α (10 ρg/ml) for 30 min. The cells were then lysed using
this. HUVECs (105 cells/150 μl medium 199) were then seeded on to lysis buffer by incubating 15 min on ice. The cell suspension was
the upper chamber in the presence and absence of harmine (0.5, 1 and centrifuged and disrupted using a syringe and centrifuged at 10000–
2 μg/ml) along with 2 ng/ml VEGF and FGF and incubated at 37 °C in 5% 11000 × g for 20 min. The crude nuclear pellet obtained was
CO2 atmosphere for 10 h. After incubation, the filters were removed, suspended in nuclear extraction buffer. Nuclei were disrupted by
fixed with methanol and stained with crystal violet. Cells migrating to using a fresh syringe, centrifuged and the supernatant was collected.
the lower surface of the polycarbonate filters were counted under a Protein concentrations of the nuclear extracts were estimated using
microscope. The results were expressed as percentage inhibition of standard Bradford method and stored at −70 °C.
invasion.
2.16.2. Transcription factor profiling
2.13. Effect of harmine on endothelial cell tube formation Transcription factor profiling was done with BD Mercury Trans-
factor kit obtained from BD Biosciences. Using an ELISA based format,
Tube formation was carried out according to the protocol of Gupta the transfactor kit detected the DNA bound transcription factors
et al. (2002). Matrigel was allowed to thaw on ice at 4 °C. A thirty (Shen et al., 2002). This method is faster, easier, and more sensitive
microlitre ice cold matrigel was pipetted into a 96-well flat bottomed than electrophoretic mobility shift assay and does not require
titre plate without introducing air bubbles and kept for 30 min at radioactivity. When nuclear extracts added to the well, DNA will
37 °C to allow gelling of matrigel. HUVECs in medium 199 were bind to their consensus sequences in the well. Bound transcription
seeded onto the layer of matrigel at a density of 1 × 103 cells/well factors in the DNA were detected by specific primary antibody
along with 2 ng/ml VEGF and FGF. Various concentrations of harmine towards NF-κBp65, NF-κBp50, NF-κB c-Rel, c-Fos, ATF-2 and CREB. A
(0.5, 1 and 2 μg/ml) were added into the wells and incubated for 48 h horse radish peroxidase-conjugated secondary antibody was then
at 37 °C in 5% CO2 atmosphere. After incubation, the cells were fixed used to detect the bound primary antibody. The enzymatic product
with Diff Quick fixative and stained with Diff Quick stain solutions. was measured with standard microtitre plate reader at 655 nm.
Tube formation was examined and photographed using an inverted Percentage inhibition was calculated by the formula, 1 − ([OD of
microscope (200×). treated/OD of control] × 100).

2.14. Effect of harmine on the microvessel outgrowth from the rat aortic 2.17. Determination of the effect of harmine on MMPs by
ring (rat aortic ring assay) gelatin zymography

The rat aortic ring assay was used as the in vitro angiogenesis study SDS-PAGE was performed with 0.1% gelatin incorporated in the
model (Zhu and Nicosia, 2002). Dorsal aorta from a freshly sacrificed separating gel (Billings et al., 1991). HUVEC cells of sub-confluent
Sprague–Dawley rat was taken out in a sterile manner and rinsed in ice cultures were pretreated with harmine (0.5, 1 and 2 mg/ml) for 24 h
cold PBS. It was then cut into ∼1 mm long pieces using surgical blades. at 37 °C in 5% CO2 atmosphere. The medium from sub-confluent (70%)
Each ring was placed in a collagen pre-coated 96-well plate. The rings bottles of treated and untreated cells were removed, cells were then
were incubated for 24 h at 37 °C in complete medium and then replaced washed and incubated in serum free medium DMEM at 37 °C for 24 h.
with conditioned medium (CM) from B16F-10 melanoma cells and The CM was then collected and subjected to zymographic analysis.
T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73 67

After determining the protein concentration, 50 μl of sample (equiv- graphed under ultraviolet light. Band intensity was measured using
alent to 100 μg protein) was activated with 5 μl trypsin solution software provided with vilber Lourmat (Photo-Capt Version 12.3 for
(75 μg/ml) in 0.1 M Tris–HCl, 10 mM CaCl2 buffer (pH-8.0) and Windows) gel documentation system and percentage expression was
incubated for 1 h at room temperature. Samples were mixed with an calculated.
equal volume of 2X sample buffer and loaded on to 11% polyacryl-
amide gels containing 0.1% gelatin. Electrophoresis was carried out at 2.19. Statistical analysis
4 °C with constant current of 2 mA/tube until the tracking dye reached
the periphery. The gels were then washed with 2% Triton X-100 in Results were expressed as mean ± S.D. The statistical analysis was
0.1 M Tris–HCl, 10 mM CaCl2 at 37 °C for 18 h followed by staining done using one-way ANOVA followed by Dunnett's test.
with Gelcode Blue stain reagent for 2 h. Gels were destained to
visualize the clear area against the dark background. 3. Results

2.18. Studies on the expression of VEGF, iNOS and COX-2 by RT-PCR 3.1. Effect of harmine on tumour specific capillary formation

Logarithmically growing B16F-10 cells (5 × 104) were incubated The number of tumour directed capillaries were significantly
for 4 h in the presence and absence of harmine at a concentration of reduced by the administration of harmine (51.32%) (Fig. 2B). The
2 μg/ml. cDNA was synthesized using the cells to-cDNA kit (Ambion harmine received group had an average of 15.33 ± 1.03 capillaries
Inc). Cells from tissue culture were washed with PBS and heated in cell whereas the control group had an average of 31.50 ± 2.81 capillaries.
lysis buffer (provided in the kit) to release the RNA into the solution. Animals treated with TNP-470 (reference compound) showed 89.42%
This was followed by a heating step to inactivate endogenous inhibition in the microvessel formation with an average number of
ribonucleases. The genomic DNA was further degraded by treating 3.33 ± 1.37 tumour directed blood capillaries (Fig. 2A).
with DNase followed by inactivation of DNase by heating at 70 °C.
Reverse transcription was performed at 42 °C for 50 min using 3.2. Effect of harmine on the production of IL-1β, IL-6, TNF-α, GM-CSF,
Moloney murine leukemia virus reverse transcriptase (supplied IL-2, VEGF and TIMP-1 during angiogenesis
along with the kit). The standard PCR products were generated by
using positive control cDNA provided in the kit. cDNA was amplified The level of serum cytokines such as IL-1β, IL-6, TNF-α, and GM-
using the following RT-PCR primer sets: VEGF (453 bp; 5′- CSF is presented in Tables 1 and 2. In control animals, the level of IL-1,
TGCTCACTTCCAGAAACACG-3′ and 5′-GGAAGGGTAAGCCACTCACA- IL-6, TNF-α and GM-CSF in the serum was drastically enhanced after
3′); iNOS (231 bp: 5′-CTTCAACACCAAGGTTGTCTG CAT-3′ and 5′- tumour challenge when compared with normal levels. Harmine
CAAGACGCGGAAACGAGTACTGTA-3′); COX-2 (256 bp; 5′-GTGGAAAA treatment effectively reduced the elevated levels of these serum
ACCTCGTCCAGA-3′ and 5′-TGATGGTGGCTGTTTTGGTA-3′); and cytokines. VEGF levels also showed an elevation in untreated control
GADPH (557 bp; 5′-CGTCCCGTAGACAAAATGGT-3′ and 5′-CCTTCCA- after tumour induction compared with the normal level. But in
CAATGCCAAAGTT-3′). Amplified PCR products were electrophoresed harmine treated animals, VEGF was significantly reduced showing
on a 1.8% agarose gel, stained with ethidium bromide, and photo- their activity on VEGF. The lowered levels of anti-angiogenic factors

Fig. 2. A) Inhibitory effect of harmine and TNP-40 on tumour-directed capillary formation. B) Quantitation of capillary formation in mice treated for 9 days with harmine, TNP40 and
untreated control. C) Viability of HUVEC following 24 h treatment with the indicated concentrations of harmine and untreated control as judged by MTT assay. Similar results were
observed in two independent experiments. ⁎⁎⁎ P b 0.001 compared to control.
68 T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73

Table 1
Effect of harmine on VEGF and pro-inflammatory cytokine profile of angiogenesis induced animals.

Cytokines Normal Control Harmine


(pg/ml)
Day 1 Day 9 Day 1 Day 9
a
VEGF 16.01 ± 4.29 71.47 ± 2.03 139.11 ± 2.17 37.63 ± 1.41 59.99 ± 1.41a
IL-1β 18.14 ± 3.75 32.13 ± 1.10 32.59 ± 1.16 31.13 ± 0.46 17.91 ± 1.48a
IL-6 36.81 ± 5.58 40.16 ± 2.57 318.86 ± 4.01 36.70 ± 2.40 73.40 ± 2.40a
TNF-α 21.18 ± 1.81 168.64 ± 1.71 582.96 ± 6.76 137.23 ± 2.34a 128.26 ± 1.71a
GM-CSF 18.12 ± 3.11 64.62 ± 0.52 27.42 ± 1.11 34.02 ± 1.61a 20.36 ± 0.52a

Blood was collected from the angiogenesis induced animals at two time points after tumour challenge. Serum was separated by centrifugation and the cytokine level was estimated
by ELISA method. All the values are mean ± S.D. (mean of triplicate).
a
P b 0.001 compared to respective control.

like IL-2 and TIMP in the control animals after tumour inoculation area were found to be 211 ± 13.5, 110 ± 18.8 and 19 ± 7.2 respectively
were significantly increased by the treatment with harmine. which was very much lower than that control where 397 ± 42.6 cells
were migrated (Fig. 3C). This concentration is nontoxic, as is evident
3.3. Effect of harmine on serum nitrite levels during angiogenesis from the MTT assay; hence, the inhibitory effect could not be
attributed to cytotoxic activity.
The serum nitrite level of control animals at 24 h after angiogen-
esis induction was slightly increased to 23.54 ± 0.50 μmoles and was 3.7. Effect of harmine on endothelial cell invasion
again elevated to 38.95 ± 0.82 μmoles on 9th day compared to normal
level (20.76 ± 0.41 μmoles). Administration of harmine could signif- Large numbers of cells were found on the lower surface of the
icantly inhibit the production of the serum nitrite level to 21.77 ± 1.13 polycarbonate membrane showing strong invasive property of HUVEC
μmoles at 24 h and to 23.21 ± 0.64 μmoles on the 9th day. through collagen in the presence of VEGF (Fig. 3B). In untreated control
1903 ± 32.5 cells were observed under the polycarbonate membrane
3.4. Effect of harmine on cell viability and it was significantly reduced to 1378.5 ± 50.2, 986.5 ± 58.7 and
335.5 ± 2.1 cells when the cell were treated with harmine at 0.5, 1, and
The percentage viability of HUVEC cells after the treatment with 2 μg/ml respectively. The percentage inhibitions in the invasion of
harmine is shown in Fig. 2C. The concentration needed for 50% HUVEC after the treatment with harmine at these concentrations were
inhibition (IC50) of growth of HUVEC cells was found to be 32.6 μg/ml. found to be 27.56%, 48.16% and 82.37% respectively. This demonstrates
The cell were 100% alive below 5 μg/ml of harmine and found to be the dose dependent inhibitory effect of harmine on the migration of
nontoxic at 0.5, 1 and 2 μg/ml of harmine and these concentrations HUVEC through collagen coated membrane which is an important step
were used for further in vitro experiments. in neo-vessel formation (Fig. 3D).

3.5. Effect of harmine on endothelial cell proliferation


3.8. Effect of harmine on endothelial cell tube formation
3
Rate of proliferation was determined by H-thymidine incorpora-
tion by the DNA of HUVECs. Thymidine incorporation is proportional Treatment of HUVECs with harmine significantly inhibited tube
to the potential of the cells to synthesize DNA. Proliferation was formation (Fig. 4A). Incubation of HUVECs on matrigel with VEGF
expressed as radioactive count per minute (cpm). HUVECs showed a resulted in the formation of elongated and tube-like structures.
very high rate of proliferation (4521 ± 155 cpm) when stimulated Harmine effectively reduced the width and length of endothelial tubes
with VEGF. Administration of harmine at non toxic concentration (0.5, at 2 μg/ml.
1, and 2 μg/ml) significantly inhibited VEGF-induced proliferation of
HUVECs in a dose dependent manner (Table 3). 3.9. Effect of harmine on rat aortic ring assay

3.6. Effect of harmine on endothelial cell motility The vessels that grow out from aortic rings are anatomically
similar to neo-vessels. Harmine at a concentration of 2 μg/ml could
The effect of harmine on the motility of HUVECs is shown in significantly inhibit the microvessel outgrowth from the rat aorta ring
Fig. 3A. Harmine significantly inhibited the VEGF-induced migration induced by the CM from the B16F10 melanoma cells (Fig. 4B-c). The
of endothelial cells in a dose dependent manner compared to control aortic rings incubated with CM from the harmine treated B16F10 cells
where HUVECs migrated into the clear area when stimulated with (pretreated CM) also showed a marked reduction in vessel out growth
VEGF. When the cells were treated with harmine at concentrations of (Fig. 4B-b) when compared with rings incubated with CM from
0.5, 1 and 2 μg/ml, the number of cells migrated towards the wound untreated B16F-10 cells (Fig. 4B-a).

Table 2
Effect of harmine on TIMP and IL-2 levels of angiogenesis induced animals.

Cytokines Normal Control Harmine


(pg/ml)
Day 1 Day 9 Day 1 Day 9
a
TIMP 600.65 ± 35.63 354.26 ± 5.26 371.34 ± 6.86 947.41 ± 4.56 1052.31 ± 61.19a
IL-2 23.21 ± 3.16 16.96 ± 0.34 18.30 ± 0.76 26.19 ± 9.52a 54.83 ± 0.41a

Blood was collected from the angiogenesis induced animals at two time points after tumour challenge. Serum was separated by centrifugation, and levels of TIMP and IL-2 were
estimated by ELISA method. All the values are mean ± S.D. (mean of triplicate).
a
P b 0.001 compared to respective control.
T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73 69

Table 3 significantly reduced this elevated level of VEGF mRNA to 9.77 ±


Effect of harmine on proliferation of HUVECs. 0.18 attomoles.
Treatment Radioactive count/min (CPM) % Inhibition

Untreated control 4521 ± 155


3.11. Effect of harmine on transcription factors
Harmine μg/ml
0.5 3614 ± 40a 20.06 Harmine significantly inhibited the translocation/activation of NF-
1 2755 ± 83a 39.06 κB subunits such as p65, p50, and c-Rel. We also found that harmine
2 1591 ± 91a 64.80
inhibited the nuclear translocation of AP-1 factors as c-fos, ATF-2, and
HUVECs (5 × 103 cells/well) were grown in 96 well flat bottom plates. After 24 h various CREB (Fig. 5A).
concentrations of harmine (0.5, 1 and 2 μg/ml) were added and incubation was
continued for 48 h. After incubation, 3H-thymidine was added to each well (1 μ Ci/well)
and incubation was continued for 18 h. Cells were lysed and radioactivity was counted
3.12. Effect of harmine on matrix metalloproteases
by using Rack Beta liquid scintillation counter. Values are mean ± S.D. (mean of
triplicate). MMP activity can be assessed using gelatin zymographic assays.
a
P b 0.001 compared to control. MMPs are generally secreted as zymogens which are activated by the
proteolytic cleavage by trypsin. As shown in Fig. 4B, harmine inhibited
the production of MMPs by HUVEC cells. Gels loaded with CM without
3.10. Quantitation of gene specific mRNA of VEGF in B16F-10 melanoma trypsin activation, did not show clear degradative areas, indicating the
cells inactive form of the enzyme (Fig. 5B-a). CM from the untreated B16F-
10 melanoma cells after the trypsin activation gave the functional
Considerable inhibition of VEGF mRNA synthesis was also observed degradative activity identical to MMP-9 (92 kDa) and MMP-2
in harmine treated melanoma cells. Untreated B16F-10 melanoma cells (72 kDa) (Fig. 5B-b). Trypsin activated CM from the untreated B16F-
showed high level of VEGF mRNA expression (26.43 ± 0.37 attomoles). 10 cells loaded gels did not show clear degradative areas when it was
Treatment with harmine at a concentration of 2 μg/ml for 4 h incubated with 10 mM EDTA (Fig. 5B-c), indicating that the enzyme

Fig. 3. Effect of harmine on in vitro angiogenesis. (A) Harmine inhibited motility of HUVEC cells toward wound area in a dose dependent manner. B) Harmine inhibited VEGF-induced
migration of HUVECs through ECM coated membrane. C) Migration was quantified by counting the cell migrated towards wound area. D) Invasion was quantified by counting the
cells that invaded to the lower side of the filter after fixation and staining and photographed at 200× magnification and% inhibiton was calculated. All the values are mean ± S.D.
(mean of triplicate). ⁎⁎⁎ P b 0.001, ⁎⁎ P b 0.01 compared to control.
70 T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73

A pre-requisite for the growth of solid tumours, vascular targeting has


been explored as a potential strategy to suppress tumour growth and
metastasis. Several angiogenic inhibitors have been developed for
targeting angiogenic factors or their receptors, extracellular matrix
compounds, and vascular endothelial cell proliferation. Among them,
thalidomide and TNP-470 are known to inhibit the growth of
endothelial cells (D'Amato et al., 1994). In addition several phytochem-
icals have been evaluated for inhibiting various components of the
mitogenic, cell survival and inflammatory signaling in tumour cells
associated with the production of angiogenic factors (Fotsis et al., 1997).
Soybean phytochemicals (Zhou et al., 1999), resveratrol from grapes
Control 2 µg/mL (Brakenhielm et al., 2001) and Andrographaloide from Andrographis
paniculata (Sheeja et al., 2007) are some of the examples.
B a The various steps in angiogenesis including degradation of the
extracellular matrix surrounding the parent vessel, migration and
proliferation of the endothelial cells and mural cells to assemble the
new vessel, lumen formation, can be studied using both in vitro and in
vivo assays. In our study when B16F-10 melanoma cells were
introduced intradermally to the mouse, tumour directed capillary
formation was increased as a tumour grows indicating a direct
correlation between the tumour survival and vessel count (Weidner
et al., 1991) Administration of harmine significantly inhibited the
b c
tumour specific new blood vessel formation in C57BL/6 mice when
they were induced angiogenesis with B16F-10 melanoma cells.
The serum VEGF level was highly elevated as quickly as 24 h after
tumour challenge, while treatment with harmine significantly inhibited
this elevation of serum VEGF level. This anti-VEGF activity of these
compounds may be due to their down regulatory action on the VEGF
mRNA expression, as is evident from the inhibition in the expression of
VEGF mRNA by B16F-10 melanoma cells upon treatment with harmine.
Fig. 4. Effect of harmine on in vitro angiogenesis. A) Harmine inhibited VEGF-induced There is considerable evidence to suggest that angiogenesis and
tube formation of HUVECs. Photographs were taken at 200× magnification. B) Harmine
inhibited VEGF-induced vessel sprouting from rat aortic ring. a) Conditioned medium
inflammation occur in an interactive and overlapping manner
(CM) alone b) CM + harmine (2 μg/ml). c) CM pretreated with harmine (2 μg/ml). (Reinders et al., 2003). Inflammatory signals regulate the expression
Experiments were repeated three times. and secretion of various angiogenic factors (Ono et al., 1999). In
pathological angiogenesis like tumour angiogenesis the autocrine,
paracrine and amphicrine interactions of the vascular endothelium
responsible for the degradation was metalloproteinase. CM from the with the pro-angiogenic and angiostatic cytokines and growth factors
harmine treated (1 and 2 μg/ml) HUVEC cells after the trypsin are irregularly regulated (Hanahan, 1997). Pro inflammatory cyto-
activation did not give any clear degraded areas (Fig. 5B-d, e), kines such as IL-1β, IL-6, TNF-α and GM-CSF contribute in the process
indicating that harmine treatment could inhibit the production of of neovascularization (Kofler et al., 2005). IL-1β has been proposed as
matrix metalloproteases. CM from the harmine treated (0.5 μg/ml) the primary driver of cell growth, proliferation, matrix degradation
HUVEC cells after the trypsin activation gave partially degraded areas and angiogenesis in invasive melanoma (Apte and Voronov, 2002;
(Fig. 5B-f), indicating that a lower dose of harmine treatment partially Dinarello, 1996). In our experiment, harmine treatment was found to
inhibited the production of matrix metalloproteases. inhibit IL-1β production in angiogenesis induced animals. Thus,
harmine mediated inhibition of IL-1β may contribute to diminished
tumour angiogenesis, probably because secretable IL-1β activates, by
3.13. Effect of harmine on the expression of VEGF, iNOS, and COX-2
autocrine/paracrine mechanisms, an inflammatory cascade in the
tumour's micro-environment. IL-6 induces transcriptional activation
Harmine treatment significantly down-regulated the expression of
of VEGF and regulates VEGF promoter activity via direct interaction
VEGF, iNOS and COX-2 transcript levels by B16F-10 melanoma cells
with STAT3. Serum IL-6 and VEGF levels were positively correlated in
compared with the untreated control (Fig. 5C). In untreated control, the
advanced cancers (Maruno et al., 1997). TNF-α enhances VEGF, IL-
percentage expression of VEGF, iNOS and COX-2 were found to be 91.7%,
8 and bFGF production in human microvascular endothelial cells and
79.2% and 52.2% respectively compared to an internal control GAPDH
induces tubular morphogenesis at the site of inflammation/tumour
(100%). Harmine treatment down-regulated the level of expression of
induction. GM-CSF is expressed in solid tumours and potentially
VEGF, iNOS and COX-2, mRNA and percentage expression were 45.2%,
enhances tumour cell proliferation and migration as well as tumour
52.3% and 15.3%, respectively compared to GAPDH (100%) of treated
angiogenesis (Bussolino et al., 1991). In this study it was found that
group (Fig. 5D). These results suggest that the anti-angiogenic effect of
the serum levels of IL-6, TNF-α, and GM-CSF in control animals were
harmine is associated with a down-regulation of pro angiogenic factors.
highly elevated after tumour induction indicating angiogenesis
induced inflammation. But harmine treatment inhibited the produc-
4. Discussion tion of these cytokines and the inhibition in these pro-inflammatory
cytokines might be the cause for the reduction of serum VEGF levels of
Tumour angiogenesis is a complex process and involves tight treated animals, since most of these pro-inflammatory cytokines are
interplay of tumour cells, endothelial cells, phagocytes and their good inducers of VEGF.
secreted factors which may act as promoters or inhibitors of IL-2 is of clinical value in natural immunity by stimulating a natural
angiogenesis (Ferrara, 2000). VEGF is the most important mitogenic killer cell and cytotoxic T-lymphocyte production (Neville et al.,
and survival factor for vascular endothelial cells. Since angiogenesis is 2001). It was reported that IL-2 presurgical immunotherapy may
T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73 71

Fig. 5. A) Effect of harmine on transcription factors: B16F-10 melanoma cells were treated with harmine (2 μg/ml) for 2 h and then incubated with TNF-α (10 pg/ml) for 30 min.
Percentage inhibition in various transcription factors were measured by ELISA method. B) Inhibitory effect of harmine on the production of matrix metalloproteinases. (a) CM from
untreated HUVECs without trypsin activation. (b) CM from untreated HUVECs with trypsin activation (c) CM from untreated HUVECs with trypsin activation + EDTA. (d) CM from
untreated HUVECs with trypsin activation + harmine (2 μg/ml). (e) CM from untreated HUVECs with trypsin activation + harmine (1 μg/ml). f) CM from untreated HUVECs with
trypsin activation + harmine (0.5 μg/ml). C) Effect of harmine VEGF-α, iNOS, and COX-2 m RNA expression by B16F-10 melanoma cells. The mRNA expression levels in each group
were normalized with GAPDH (house-keeping gene). D) Band intensity was measured using software provided with vilber Lourmat gel documentation system and percentage
expression of mRNA was calculated.

counteract surgery-induced stimulation of the angiogenesis, by either several types of cancer cells and is associated with the suppression of
opposing the decline in blood levels of the anti-angiogenetic cytokine angiogenesis, invasion and metastasis in cancer (Huang et al., 1999).
IL-12, or reducing the increase in those of the angiogenic factor VEGF. One of the genes expressed as a result of NFκB signaling in
Therefore, the control of VEGF secretion could influence the efficacy of angiogenesis is the prostaglandin synthesis enzyme, COX-2. Impor-
IL-2 (Brivio et al., 2002). In the present investigation, we found that tantly, the COX-2 enzyme is involved in the induction of VEGF and
administration of harmine significantly increased IL-2 levels, which FGF-2 growth factors that stimulate angiogenesis (Boosani et al.,
may contribute to the stimulation of the immune system against the 2007). COX-2 expression is also induced by pro inflammatory and
tumour growth by VEGF-induced angiogenesis. TIMP inhibits the mitogenic stimuli such as growth factors (VEGF) and cytokines (TNF-
activities of MMP-1, MMP-2 and MMP-9, which leads to reduced ECM α and IL1-β). COX-2 has also been implicated as playing roles in
remodeling and suppression of endothelial cell migration and endothelial cells spreading and migration (Dormond et al., 2002; Zaric
invasion (Gomez et al., 1997b). TIMP-1 was increased in the harmine and Ruegg, 2005). Vasodilation, by smooth muscle relaxation,
administered group suggesting that increased endogenous TIMP-1 mediated by nitric oxide is a pre-requisite for endothelial cells to
could inhibit MMP activity and consequently inhibit ECM remodeling. enter angiogenic cascade (Griffioen and Molema, 2000). Since COX-2
Similar to other pro-angiogenic factors, intradermal introduction of and iNOS play important roles in the progression of various types of
highly metastatic B16F-10 melanoma cells resulted in drastic eleva- cancer by stimulating angiogenesis, invasion, and apoptosis (Cuezva
tion of serum nitrite level of untreated angiogenesis induced animals, et al., 2002; Gasparini et al., 2003), we tested whether harmine blocks
which may be the reason for increased tumour directed capillary the activity of these molecules. Expression analysis clearly demon-
formation in them. But the administration of harmine significantly strated that treatment of B16F-10 cells with harmine could signifi-
reduced the elevated serum nitrite level after tumour challenge, cantly down-regulate the production of iNOS and COX-2.
which in turn resulted in decreased tumour directed capillary To test the anti-angiogenic activity effects of harmine in vitro, we
formation. treated HUVEC cells with harmine and analyzed various steps in
Inflammation-driven expression of an angiogenic factor can also angiogenesis. Assays that allow measurement of endothelial cell
be mediated by the activation of NF- B signaling (Bhat and Singh, motility in response to added factors include the proliferation, migration
2008). VEGF is one of the most potent angiogenic factors that can be assay (Scrape wound), invasion assay (transwell), etc. (Lampugnani,
regulated by activated NF- B, because its genes have a B binding site 1999). Effects of harmine on cell survival and proliferation were
in its promoter region (Kang et al., 2005; Karin, 2006). Inhibition of measured by direct cell counting MTT assay and thymidine incorpora-
NF- B activity leads to decreased expression of VEGF and IL-8 in tion assay. It was found that the rate of proliferation was decreased in a
72 T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73

dose dependent manner. Because of the prime role of MMP in Chen, Q., Chao, R., Chen, H., Hou, X., Yan, H., Zhou, S., Peng, W., Xu, A., 2005. Antitumor
and neurotoxic effects of novel harmine derivatives and structure-activity
endothelial cell invasion and migration, the inhibition of the activation relationship analysis. Int. J. Cancer 114, 675–682.
of MMPs by harmine resulted in the significant reduction of endothelial Cole, S.P., 1986. Rapid chemosensitivity testing of human lung tumor cells using the
cell motility and invasion though collagen matrix in response to VEGF in MTT assay. Cancer Chemother. Pharmacol. 17, 259–263.
Cuezva, J.M., Krajewska, M., de Heredia, M.L., Krajewski, S., Santamaria, G., Kim, H.,
a dose dependent manner. The tube formation stage of angiogenesis can Zapata, J.M., Marusawa, H., Chamorro, M., Reed, J.C., 2002. The bioenergetic
be modeled in vitro by plating endothelial cells with matrigel (Kubota et signature of cancer: a marker of tumor progression. Cancer Res. 62, 6674–6681.
al., 1988). The formation of tubes on matrigel involves endothelial cell D'Amato, R.J., Loughnan, M.S., Flynn, E., Folkman, J., 1994. Thalidomide is an inhibitor of
angiogenesis. Proc. Natl Acad. Sci. USA 91, 4082–4085.
attachment, migration and production of enzymes capable of remodel- Dignam, J.D., Lebovitz, R.M., Roeder, R.G., 1983. Accurate transcription initiation by RNA
ing ECM. It was shown that a reduction in tube formation by harmine polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids
treated endothelial cells was associated with a decrease in the Res. 11, 1475–1489.
Dinarello, C.A., 1996. Biologic basis for interleukin-1 in disease. Blood 87, 2095–2147.
proteolytic activities of both MMP-2 and MMP-9. The aortic ring assay
Dormond, O., Bezzi, M., Mariotti, A., Ruegg, C., 2002. Prostaglandin E2 promotes integrin
is an organ culture model in which angiogenic vessels grow from a alpha Vbeta 3-dependent endothelial cell adhesion, rac-activation, and spreading
segment of the aorta (Masson et al., 2002). The vessels that grow out through cAMP/PKA-dependent signaling. J. Biol. Chem. 277, 45838–45846.
from aortic rings are anatomically similar to neovessels in vivo Dvorak, H.F., 2005. Angiogenesis: update 2005. J. Thromb. Haemost. 3, 1835–1842.
Feldman, A.L., Libutti, S.K., 2000. Progress in antiangiogenic gene therapy of cancer.
(Goodwin, 2007). A marked decrease in vessel out growth from the Cancer 89, 1181–1194.
rat aortic ring was observed in harmine treated condition medium. This Ferrara, N., 2000. VEGF: an update on biological and therapeutic aspects. Curr. Opin.
may be due to the absence of pro-angiogenic factors in CM. The effect of Biotechnol. 11, 617–624.
Fotsis, T., Pepper, M.S., Aktas, E., Breit, S., Rasku, S., Adlercreutz, H., Wahala, K.,
harmine on MMPs was confirmed by gelatin zymographic analysis and Montesano, R., Schweigerer, L., 1997. Flavonoids, dietary-derived inhibitors of cell
it was clearly revealed that harmine inhibited the production of MMP by proliferation and in vitro angiogenesis. Cancer Res. 57, 2916–2921.
HUVEC cells in vitro. Gasparini, G., Longo, R., Sarmiento, R., Morabito, A., 2003. Inhibitors of cyclo-oxygenase
2: a new class of anticancer agents? Lancet Oncol. 4, 605–615.
In conclusion, we present experimental evidence suggesting that Gomez, D.E., Alonso, D.F., Yoshiji, H., Thorgeirsson, U.P., 1997a. Tissue inhibitors of
harmine exerts multiple effects on the inhibition of tumour metalloproteinases: structure, regulation and biological functions. Eur. J. Cell Biol.
angiogenesis and it's mechanism of anti-angiogenesis is pleotropic. 74, 111–122.
Gomez, M., Luque, A., del Pozo, M.A., Hogg, N., Sanchez-Madrid, F., Cabanas, C., 1997b.
Our data clearly shows that harmine considerably inhibits tumour Functional relevance during lymphocyte migration and cellular localization of
directed neo-vessel formation by inhibiting pro-angiogenic and pro- activated beta1 integrins. Eur. J. Immunol. 27, 8–16.
inflammatory factors in vivo and in vitro at relatively lower Goodwin, A.M., 2007. in vitro assays of angiogenesis for assessment of angiogenic and
anti-angiogenic agents. Microvasc. Res. 74, 172–183.
concentrations so harmine may be used as anti-tumour agent by
Green, L.C., Wagner, D.A., Glogowski, J., Skipper, P.L., Wishnok, J.S., Tannenbaum, S.R.,
targeting angiogenesis and inflammation. 1982. Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal.
Biochem. 126, 131–138.
Griffioen, A.W., Molema, G., 2000. Angiogenesis: potentials for pharmacologic
Acknowledgment intervention in the treatment of cancer, cardiovascular diseases, and chronic
inflammation. Pharmacol. Rev. 52, 237–268.
Guo, H.B., Lee, I., Kamar, M., Akiyama, S.K., Pierce, M., 2002. Aberrant N-glycosylation of
We acknowledge Dr. Ramadasan Kuttan, Research Director, Amala beta1 integrin causes reduced alpha5beta1 integrin clustering and stimulates cell
Cancer Research Centre, for his valuable suggestions and support in migration. Cancer Res. 62, 6837–6845.
Gupta, K., Kshirsagar, S., Chang, L., Schwartz, R., Law, P.Y., Yee, D., Hebbel, R.P., 2002.
this study. Morphine stimulates angiogenesis by activating proangiogenic and survival-promot-
ing signaling and promotes breast tumor growth. Cancer Res. 62, 4491–4498.
Hanahan, D., 1997. Signaling vascular morphogenesis and maintenance. Science 277,
References 48–50.
Huang, Y.T., Hwang, J.J., Lee, P.P., Ke, F.C., Huang, J.H., Huang, C.J., Kandaswami, C.,
Abe, A., Yamada, H., 2009. Harmol induces apoptosis by caspase-8 activation Middleton Jr., E., Lee, M.T., 1999. Effects of luteolin and quercetin, inhibitors of
independently of Fas/Fas ligand interaction in human lung carcinoma H596 cells. tyrosine kinase, on cell growth and metastasis-associated properties in A431 cells
Anticancer Drugs 20, 373–381. overexpressing epidermal growth factor receptor. Br. J. Pharmacol. 128, 999–1010.
Adams, R.H., Alitalo, K., 2007. Molecular regulation of angiogenesis and lymphangio- Ishida, J., Wang, H.K., Bastow, K.F., Hu, C.Q., Lee, K.H., 1999. Antitumor agents 201.
genesis. Nat. Rev. Mol. Cell Biol. 8, 464–478. Cytotoxicity of harmine and beta-carboline analogs. Bioorg. Med. Chem. Lett. 9,
Albini, A., Iwamoto, Y., Kleinman, H.K., Martin, G.R., Aaronson, S.A., Kozlowski, J.M., 3319–3324.
McEwan, R.N., 1987. A rapid in vitro assay for quantitating the invasive potential of Jaffe, E.A., Nachman, R.L., Becker, C.G., Minick, C.R., 1973. Culture of human endothelial
tumor cells. Cancer Res. 47, 3239–3245. cells derived from umbilical veins. Identification by morphologic and immunologic
Apte, R.N., Voronov, E., 2002. Interleukin-1 — a major pleiotropic cytokine in tumor- criteria. J. Clin. Invest. 52, 2745–2756.
host interactions. Semin. Cancer Biol. 12, 277–290. Kang, S., Kim, Y.B., Kim, M.H., Yoon, K.S., Kim, J.W., Park, N.H., Song, Y.S., Kang, D., Yoo, K.Y.,
Arshad, N., Zitterl-Eglseer, K., Hasnain, S., Hess, M., 2008. Effect of Peganum harmala or Kang, S.B., Lee, H.P., 2005. Polymorphism in the nuclear factor kappa-B binding
its beta-carboline alkaloids on certain antibiotic resistant strains of bacteria and promoter region of cyclooxygenase-2 is associated with an increased risk of bladder
protozoa from poultry. Phytother. Res. 22, 1533–1538. cancer. Cancer Lett. 217, 11–16.
Berrougui, H., Martin-Cordero, C., Khalil, A., Hmamouchi, M., Ettaib, A., Marhuenda, E., Karin, M., 2006. Nuclear factor-kappaB in cancer development and progression. Nature
Herrera, M.D., 2006. Vasorelaxant effects of harmine and harmaline extracted from 441, 431–436.
Peganum harmala L. seeds in isolated rat aorta. Pharmacol. Res. 54, 150–157. Kofler, S., Nickel, T., Weis, M., 2005. Role of cytokines in cardiovascular diseases: a focus
Bhat, T.A., Singh, R.P., 2008. Tumor angiogenesis — a potential target in cancer on endothelial responses to inflammation. Clin. Sci. (Lond) 108, 205–213.
chemoprevention. Food Chem. Toxicol. 46, 1334–1345. Kubota, Y., Kleinman, H.K., Martin, G.R., Lawley, T.J., 1988. Role of laminin and basement
Billings, P.C., Habres, J.M., Liao, D.C., Tuttle, S.W., 1991. Human fibroblasts contain a membrane in the morphological differentiation of human endothelial cells into
proteolytic activity which is inhibited by the Bowman–Birk protease inhibitor. capillary-like structures. J. Cell Biol. 107, 1589–1598.
Cancer Res. 51, 5539–5543. Lal, B.K., Varma, S., Pappas, P.J., Hobson 2nd, R.W., Duran, W.N., 2001. VEGF increases
Boosani, C.S., Mannam, A.P., Cosgrove, D., Silva, R., Hodivala-Dilke, K.M., Keshamouni, V.G., permeability of the endothelial cell monolayer by activation of PKB/akt, endothelial
Sudhakar, A., 2007. Regulation of COX-2 mediated signaling by alpha3 type IV nitric-oxide synthase, and MAP kinase pathways. Microvasc. Res. 62, 252–262.
noncollagenous domain in tumor angiogenesis. Blood 110, 1168–1177. Lampugnani, M.G., 1999. Cell migration into a wounded area in vitro. Methods Mol. Biol.
Brakenhielm, E., Cao, R., Cao, Y., 2001. Suppression of angiogenesis, tumor growth, and 96, 177–182.
wound healing by resveratrol, a natural compound in red wine and grapes. FASEB J. Leyon, P.V., Kuttan, G., 2004. Effect of Tinospora cordifolia on the cytokine profile of
15, 1798–1800. angiogenesis-induced animals. Int. Immunopharmacol. 4, 1569–1575.
Brivio, F., Lissoni, P., Rovelli, F., Nespoli, A., Uggeri, F., Fumagalli, L., Gardani, G., 2002. Maruno, M., Kovach, J.S., Kelly, P.J., Yanagihara, T., 1997. Distribution of endogenous
Effects of IL-2 preoperative immunotherapy on surgery-induced changes in tumour necrosis factor alpha in gliomas. J. Clin. Pathol. 50, 559–562.
angiogenic regulation and its prevention of VEGF increase and IL-12 decline. Masson, V.V., Devy, L., Grignet-Debrus, C., Bernt, S., Bajou, K., Blacher, S., Roland, G.,
Hepatogastroenterology 49, 385–387. Chang, Y., Fong, T., Carmeliet, P., Foidart, J.M., Noel, A., 2002. Mouse aortic ring
Bussolino, F., Ziche, M., Wang, J.M., Alessi, D., Morbidelli, L., Cremona, O., Bosia, A., assay: a new approach of the molecular genetics of angiogenesis. Biol. Proced.
Marchisio, P.C., Mantovani, A., 1991. in vitro and in vivo activation of endothelial Online 4, 24–31.
cells by colony-stimulating factors. J. Clin. Invest. 87, 986–995. Moura, D.J., Richter, M.F., Boeira, J.M., Pegas Henriques, J.A., Saffi, J., 2007. Antioxidant
Campling, B.G., Pym, J., Baker, H.M., Cole, S.P., Lam, Y.M., 1991. Chemosensitivity testing properties of beta-carboline alkaloids are related to their antimutagenic and
of small cell lung cancer using the MTT assay. Br. J. Cancer 63, 75–83. antigenotoxic activities. Mutagenesis 22, 293–302.
T.P. Hamsa, G. Kuttan / European Journal of Pharmacology 649 (2010) 64–73 73

Neville, M.E., Robb, R.J., Popescu, M.C., 2001. In situ vaccination against a non- Shen, Z., Peedikayil, J., Olson, G.K., Siebert, P.D., Fang, Y., 2002. Multiple transcription
immunogenic tumour using intratumoural injections of liposomal interleukin-2. factor profiling by enzyme-linked immunoassay. Biotechniques 32(5):1168,
Cytokine 16, 239–250. 1170-2, 1174 passim.
Ono, M., Torisu, H., Fukushi, J., Nishie, A., Kuwano, M., 1999. Biological implications of Weidner, N., Semple, J.P., Welch, W.R., Folkman, J., 1991. Tumor angiogenesis and
macrophage infiltration in human tumor angiogenesis. Cancer Chemother. metastasis — correlation in invasive breast carcinoma. N Engl J. Med. 324, 1–8.
Pharmacol. 43 (Suppl), S69–S71. Zaric, J., Ruegg, C., 2005. Integrin-mediated adhesion and soluble ligand binding
Reinders, M.E., Sho, M., Izawa, A., Wang, P., Mukhopadhyay, D., Koss, K.E., Geehan, C.S., stabilize COX-2 protein levels in endothelial cells by inducing expression and
Luster, A.D., Sayegh, M.H., Briscoe, D.M., 2003. Proinflammatory functions of preventing degradation. J. Biol. Chem. 280, 1077–1085.
vascular endothelial growth factor in alloimmunity. J. Clin. Invest. 112, 1655–1665. Zhou, J.R., Gugger, E.T., Tanaka, T., Guo, Y., Blackburn, G.L., Clinton, S.K., 1999. Soybean
Roberts, W.G., Palade, G.E., 1997. Neovasculature induced by vascular endothelial phytochemicals inhibit the growth of transplantable human prostate carcinoma
growth factor is fenestrated. Cancer Res. 57, 765–772. and tumor angiogenesis in mice. J. Nutr. 129, 1628–1635.
Sethi, G., Sung, B., Aggarwal, B.B., 2008. TNF: a master switch for inflammation to Zhu, W.H., Nicosia, R.F., 2002. The thin prep rat aortic ring assay: a modified method for
cancer. Front. Biosci. 13, 5094–5107. the characterization of angiogenesis in whole mounts. Angiogenesis 5, 81–86.
Sheeja, K., Guruvayoorappan, C., Kuttan, G., 2007. Antiangiogenic activity of Zhu, W.H., Guo, X., Villaschi, S., Francesco Nicosia, R., 2000. Regulation of vascular
Andrographis paniculata extract and andrographolide. Int. Immunopharmacol. 7, growth and regression by matrix metalloproteinases in the rat aorta model of
211–221. angiogenesis. Lab. Investig. 80, 545–555.

You might also like