Hann Chu and Yang10.1073/pnas.0611681104XXYYYYY103.

Supporting Information

Files in this Data Supplement:

SI Figure 6
SI Table 2
SI Table 3
SI Figure 7
SI Materials and Methods




SI Figure 6

Fig. 6. Flow-chart for assays and infection experiments. (A) For the primary assay, cells were infected with DENV at an moi of 1 incubation with virus for 60 min (37°C, 5% CO2) with gentle rocking. Following this, they were washed to remove excess virus. Culture medium containing the test compounds at set concentrations were then added to the cells. Following 3 days (37°C, 5% CO2), cells were fixed and then stained for the presence of DENV Envelope protein (Env). (B) To evaluate compounds for the ability to inhibit postviral entry processes ("postinoculation"), cells were infected with DENV at moi 1 and then treated with test compound as described above. On day 3 postinfection, culture supernatants were harvested and viral titers were quantified by plaque assay. (C) To evaluate compounds for the ability to inhibit processes that occur early in the viral life cycle ("preinoculation"), cells were preincubated with compound for 2 h (37°C, 5% CO2). Following this, three washes were performed to remove excess compound. Cells were then infected with DENV as described above. On day 3, postinfection, culture supernatants were harvested and viral titers were quantified by plaque assay. (D) In plaque assays, test samples were used to generate a dilution series that was then used to infect cells for 1 h (37°C, 5% CO2) with gentle rocking. After washes to remove excess virus, the cell monolayers were covered with a carboxymethylcellulose (CMC) overlay and incubated at 37°C (5% CO2) for 5 days. Cells were then fixed and stained with crystal violet, and plaques were quantified to determine titers (per ml of supernatant) in the test samples.





Fig. 7. Anti-DENV activity of AZD0530. (A) AZD0530, a Src-selective kinase inhibitor, decreases the productive viral titer in DENV2-infected C6/36 cells, Vero cells, and Huh7 cells in a dose-dependent manner. (B) DENV viral envelope protein accumulates in AZD0530-treated Vero cells. DENV-infected Vero cell monolayers were treated with 2.5 µM AZD0530 or DMSO. Noninfected cells are indicated by arrowheads; the accumulation of viral envelope protein in the perinuclear region is indicated by arrows. Cell nuclei are stained blue with DAPI.





Table 2. List of cellular protein kinases targeted by the inhibitors screened in this study

Cellular kinase

Type of kinase

c-Abl

Tyrosine kinase

Akt/PKB

Serine/threonine kinase

Aurora

Serine/threonine kinase

Bmx

Serine/threonine kinase

Casein kinase

Serine/threonine kinase

Cdk1/cyclin B

Serine/threonine kinase

Cdk2/cyclin A

Serine/threonine kinase

Cdk2/cyclin E

Serine/threonine kinase

Cdk3/cyclin E

Serine/threonine kinase

Cdk4/cyclin D1

Serine/threonine kinase

Cdk5/cyclin p25

Serine/threonine kinase

Chk 1/2

Serine/threonine kinase

DYRK1a

Tyrosine kinase

EGFR

Tyrosine kinase

Eph

Tyrosine kinase

FGFR1-4

Tyrosine kinase

Flt1/3

Tyrosine kinase

Fms/CSF-1R

Tyrosine kinase

GSKα/β

Serine/threonine kinase

IKK

Serine/threonine kinase

IRAK

Serine/threonine kinase

JAK1-3

Tyrosine kinase

JNK/SAPK

Serine/threonine kinase

KDR

Tyrosine kinase

c-Kit

Tyrosine kinase

Lck

Tyrosine kinase

LIM kinase

Serine/threonine kinase

MAP kinase/Erk

Serine/threonine kinase

MEK1/2

Serine/threonine kinase

c-Met

Tyrosine kinase

p38/SAPK

Serine/threonine kinase

PDGFR

Tyrosine kinase

PI 3-kinase

Serine/threonine kinase

PKC and isoforms

Serine/threonine kinase

PKR

Serine/threonine kinase

Raf-1

Serine/threonine kinase

B-Raf

Serine/threonine kinase

ROK/ROCK

Serine/threonine kinase

c-Src

Tyrosine kinase

Syk

Tyrosine kinase

TGF-β R1 kinase

Serine/threonine kinase

Trk A/B

Tyrosine kinase

Tpl2/Cot

Serine/threonine kinase

VEGFR2

Tyrosine kinase





Table 3. Structure and specificity of inhibitors examined in secondary screening efforts

Code

Other names

Source

Structure

Reference

K002

NA

Synthesis

 

(N. Gray, personal communication)

K003

NA

Synthesis

(1)

K005

Dasatinib

Synthesis

(2)

K013

GNF-2

Synthesis

(3)

K014

Imatinib

Synthesis

(4)

K025

SU11652

CBC572660

(5)

K026

SU5271

CBC234490

(6)

K028

Lavendustin A

CBC428150

(7)

K030

NA

CBC567805

(8)

K032

Tetrabromobenzotriazole

CBC218697

(9)

K039

NA

CBC553008

(10)

K040

NA

CBC420099

(11)

K045

AZD0530

Synthesis

(12)

K114

Kenpaullone

CBC422000

(13)

K115

Lavendustin C

CBC234450

(14)

K116

MC7-HCl,

CBC475880

(15)

K117

NA

CBC574711

(16)

K118

Tyrphostin 46

BioMol-EI257

(17)

1. Okram B, Nagle A, Adrian FJ, Lee C, Ren P, Wang X, Sim T, Xie Y, Wang X, Xia G, et al. (2006) Chem Biol 13:779-786.

2. Shah NP, Tran C, Lee FY, Chen P, Norris D, Sawyers CL (2004) Science 305:399-401.

3. Adrian FJ, Ding Q, Sim T, Velentza A, Sloan C, Liu Y, Zhang G, Hur W, Ding S, Manley P, et al. (2006) Nat Chem Biol 2:95-102.

4. Buchdunger E, Zimmermann J, Mett H, Meyer T, Muller M, Druker BJ, Lydon N B (1996) Cancer Res 56:100-104.

5. Sun L, Liang C, Shirazian S, Zhou Y, Miller T, Cui J, Fukuda J. Y, Chu J. Y, Nematalla A, Wang X, et al. (2003) J Med Chem 46:1116-1119.

6. Fry DW, Kraker AJ, McMichael A, Ambroso LA, Nelson JM, Leopold WR, Connors RW, Bridges AJ (1994) Science 265:1093-1095.

7. Bishop R, Martinez R, Nakamura KD, Weber MJ (1983) Biochem Biophys Res Commun 115:536-543.

8. Tian G, Cory M, Smith AA, Knight WB (2001) Biochemistry 40:7084-7091.

9. Sarno S, Reddy H, Meggio F, Ruzzene M, Davies SP, Donella-Deana A, Shugar D, Pinna LA (2001) FEBS Lett 496:44-48.

10. Lackey K, Cory M, Davis R, Frye SV, Harris PA, Hunter RN, Jung DK, McDonald OB, McNutt RW, Peel MR, et al. (2000) Bioorg Med Chem Lett 10:223-226.

11. Thompson JE, Cubbon RM, Cummings RT, Wicker LS, Frankshun R, Cunningham BR, Cameron PM, Meinke PT, Liverton N, Weng Y, DeMartino JA (2002) Bioorg Med Chem Lett 12:1219-1223.

12. Hennequin LF, Allen J, Breed J, Curwen J, Fennell M, Green TP, Lambert-van der Brempt C, Morgentin R, Norman RA, Olivier A, et al. (2006) J Med Chem 49:6465-6488.

13. Schultz C, Link A, Leost M, Zaharevitz DW, Gussio R, Sausville EA, Meijer L, Kunick C (1999) J Med Chem 42:2909-2919.

14. Onoda T, Iinuma H, Sasaki Y, Hamada M, Isshiki K, Naganawa H, Takeuchi T, Tatsuta K, Umezawa K (1989) J Nat Prod 52:1252-1257.

15. Saitoh M, Ishikawa T, Matsushima S, Naka M, Hidaka H (1987) J Biol Chem 262:7796-7801.

16. Lai JY, Cox PJ, Patel R, Sadiq S, Aldous DJ, Thurairatnam S, Smith K, Wheeler D, Jagpal S, Parveen S, et al. (2003) Bioorg Med Chem Lett 13:3111-3114.

17. Smyth MS, Stefanova I, Hartmann F, Horak ID, Osherov N, Levitzki A, Burke TR, Jr (1993) J Med Chem 36:3010-3014.





SI Materials and Methods

Cell lines, Virus and Antibodies. All work with infectious dengue virus was performed in a biosafety level 2+ laboratory using protocols approved by the Harvard Committee on Microbiological Safety. The human hepatoma Huh-7 cell line and green African monkey kidney (Vero) cells were cultured in DMEM medium with 10% FBS at 37ºC with 5% CO2. C6/36 cells, a continuous mosquito cell line derived from Aedes albopictus (Diptera: Culicidae) embryonic tissue was grown in L-15 medium containing 10 % FBS at 28 ºC. Both Huh-7 and Vero cells were purchased from ATCC; C6/36 cells were a kind gift from Lee Gerhke (Massachusetts Institute of Technology, Cambridge, MA). The four serotypes of DENV (DENV1, Hawaii strain; DENV2, New Guinea C strain; DENV3, H87 strain; and DENV4, H241 strain) and Modoc virus (M544 strain) used in this study were propagated in C6/36 cells throughout this study. DENV serotypes 1, 3, and 4, and Modoc virus were purchased from ATCC (Manassas, VA). DENV2 and poliovirus type 1 were provided by Lee Gerhke (Massachusetts Institute of Technology) and Marylynn V. Yates (University of California, Riverside, CA), respectively. Poliovirus type 1 was cultured in Vero cells. Quantification of the viruses was performed with Vero cells using a plaque assay as described in the following section. Monoclonal anti-DENV E protein antibody (US Biologicals, Swampscott, MA), polyclonal anti-DENV E protein antibody (Abcam, Cambridge, MA), polyclonal anti-Modoc virus antibody (ATCC, Manassas, VA), and monoclonal anti-poliovirus type 1 antibody (Chemicon, Temecula, CA) were used for immunofluorescence detection of virus infection in cells. Antibodies against c-Src and GAPDH, respectively, were purchased from Abcam (Cambridge, MA).

Immunofluorescence Assay for Dengue Virus Infection.

Using standard protocols, Vero cells (ATCC, Manassas, VA) were harvested from a T75 culture flask. The trypsinized cells were centrifuged at 300 ´ g for 5 min, and the cell pellet was suspended in DMEM (Cellgro, Herndon, VA) with 10% FBS (Cellgro, Herndon, VA). Cells were then seeded at 2,000 cells per well in the 384-well plate (Corning, NY). The cells were incubated at 37°C with 5% CO2 in a humidifying incubator for 16 h. The cell culture medium was gently aspirated from the wells. Cells were washed thrice with 50 µl of PBS (Cellgro, Herndon, VA) per well. Fifty microliters of the dengue virus supernatant (adjusted to a multiplicity of infection of 1) were then transferred into each well of the 384-well plate. All infections were carried out in a BL2 biosafety cabinet. The virus infection was carried out for 1 h in a 37°C, 5% CO2, humidifying incubator, with gentle rocking every 15 min. The virus suspension was gently aspirated from each of the wells, and the wells were washed thrice with 50 µl of PBS (Cellgro, Herndon, VA) per well. One hundred microliters of maintenance medium (DMEM with 2 % FBS, Cellgro, Herndon, VA) was then aliquotted into each well of the 384-well plate. The cells were incubated for 3 days in a humidifying incubator at 37°C with 5% CO2 before proceeding to immunofluorescence staining.

The cells were first washed once with 50 µl of PBS (Cellgro, Herndon, VA) per well in the 384-well format. The cells were then fixed with 50 µl of cold absolute methanol (Sigma, St. Louis, MO) per well in the 384-well format for 15 min at -20ºC. The subsequent cell washing steps were carried out using an automated 384-well format plate washer (EMBLA, Molecular Devices, Sunnyvale, CA). The cells were washed and rehydrated with PBS (Cellgro, Herndon, VA) by pipetting 50 µl of PBS per well in the 384-well format for 10 min. To prevent nonspecific binding of antibodies, a blocking step using 1% BSA (Sigma, St. Louis, MO) in PBS for 1 h at room temperature was conducted. Thirty microliters of a 1:5,000 diluted anti-DENV2 virus envelope antibody (US Biologicals, Swampscott, MA) was added to each well, followed by incubation of the plate for 1 h at 37 ºC. Following this, the cells were washed three times with PBS (Cellgro, Herndon, VA) with 0.05% Trition-X 100 (Sigma, St. Louis, MO) per well in the 384-well format (10 minutes/wash). Goat anti-murine monoclonal-FITC antibody (Jackson Immunoresearch, WestGrove, PA) diluted 1:5,000 was then added to each well. Incubation was carried out for 1 h at 37 ºC. After the incubation, the cells were again washed three times with PBS (Cellgro, Herndon, VA) with 0.05% Trition-X 100 (Sigma, St. Louis, MO). The cells were then counter-stained with DAPI (100 nM; Molecular Probes) prior to collection of image data by the immunofluorescence microscopy with the excitation wavelength (495 nm) and emission wavelength (520 nm) for FITC and the excitation wavelength (358 nm) and emission wavelength (461 nm) for DAPI.

Data Acquisition.

The immunofluorescence images of the stained cells were acquired using an ImageXpress automated fluorescence microscope system (Molecular Devices, Sunnyvale, CA). Data collection and autofocusing parameters were preset using MetaExpress program (Molecular Devices, Sunnyvale, CA). Under the automated image capturing function, each well of the 384-well format was divided into 4 sectors and a single field of image was captured per sector, yielding four images per well. A generic segmentation tool function was used to identify the two different stains with intensities above background staining and data collection was obtained by logging the measurements. Data analysis after image acquisition was carried out using the cell sorting application modules (Molecular Devices, Sunnyvale, CA). DAPI-stained nuclei were counted to determine the total cell populations while FITC-stained cytoplasm was scored to determine the number of virus-infected cells. Any images with less than 500 cells were excluded from data analysis by the cell sorting module. To calculate the Z' factor (statistical measurement of the distance between the standard deviations for the signal versus the noise of an assay), 100 wells of a 384-well plate are treated with 6 μg/ml MPA in 1% final DMSO, and 100 wells were treated with 1% DMSO. The Z' factor is calculated by the equation:

1 − (3 Ã— S.D.DMSO + 3 Ã— S.D.MPA)/(meanDMSO − meanMPA).

Infectious Virus Plaque Assay.

Vero cells (ATCC) were harvested from a 75 cm2 culture flask. The trypsinized cells were centrifuged at 300 x g for five minutes and the cell pellet was suspended in DMEM (Cellgro, Herndon, VA) with 10% FBS (Cellgro, Herndon, VA). Cells were then seeded at 5 ´ 104 cells per well in the 24-well plate (Corning, NY). The cells were incubated at 37°C with 5% CO2 in a humidifying incubator overnight. The cell culture media were gently aspirated from the wells and washed thrice with 0.5 ml of PBS (Cellgro, Herndon, VA) per well. All virus infections were carried out in a BL2 biosafety cabinet. Ten-fold serial dilution of the virus sample was prepared in virus diluent (Earle's balanced salt buffer, Cellgro, Herndon, VA). Aliquot of 0.1 ml from the appropriate dilutions were inoculated in triplicate of a sterile 24-well plate (corning, NY) and incubated for 1 hour at 37°C with rocking every 15 min. After 1 h of adsorption, the excess inocula were removed by washing thrice of each well with 0.5 ml of PBS (Cellgro, Herndon, VA). Two milliliters of 2 % w/v carboxymethylcellulose (Sigma, St. Louis, MO) in DMEM with 2% FBS is layered onto the infected monolayers. The plates were incubated in a humidified incubator at 37°C with 5% CO2 for 5 days. Dengue virus plaques are visualized by staining the monolayer with 0.5 % crystal violet (sigma) / 25 % formaldehyde solution (VWR, Westchester, PA) for 30 minutes at room temperature and washed off with water. The plates were allowed to dry before counting the virus plaques.

Cytotoxicity Assays

. The cytotoxicity of each of the small molecule inhibitors used in this study was assessed by incubating Vero cells with an eight point dilution series of each protein kinase inhibitor (concentration range of 10 µM-0.078 µM) in culture medium in 384-well plates for 3 days, corresponding to the incubation period of cells with compounds in the primary screen. Following this, cell viability was assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT assay, Chemicon, Temecula, CA) according to the manufacturer's recommendations.

siRNA Experiments

We elected to use a SMARTpool (Dharmacon, Chicago, IL) of four individual siRNAs directed against c-Src since such pools have been previously demonstrated to produce fewer off-target effects than equivalent concentrations of single or paired siRNAs. Prior to any work with DENV-infected cells, we determined the concentration of the SMARTpool required to achieve significant knock-down of c-Src expression. These results are partially exhibited in Figure 3A, which shows that c-Src levels are reduced at 200 nM SMARTpool but that significant c-Src remains. In our experiments, 300 nM SMARTpool was required to achieve knockdown of the majority of c-Src activity. Consequently, we evaluated the DENV titer produced in cells treated with 200 nM and 300 nM concentrations of the SMARTpool reagents directed against c-Src, siGlo (negative control), or GAPDH (negative control) and compared those to the DENV titers produced in untreated cells. These experimental conditions are consistent with other published reports utilizing the Dharmacon SMARTpool (Chicago, IL) for "knockdown" of c-Src expression .

SDS-PAGE Electrophoresis and Western Analysis.

A 10 % separation gel in 0.25 M Tris-HCl pH 8.8, was cast between two grease-free glass plates (BioRad, Hercules, CA). A 4.5% Stacking gel in 0.125 M Tris-HCl pH 6.8, was then layered over the separation gel with comb in position and allowed to polymerize. The comb was removed and the wells were flushed with Tris-glycine electrophoresis buffer (196mM glycine / 0.1% SDS / 50mM Tris•HCl, pH 8.3) before loading the samples. 15 ml of each samples per well was used. Prior to loading, each protein samples was boiled for 1 min with 5 ml of glycerol-bromophenol blue dye and dithioerytritol mixture. Five ml of the prestained molecular weight marker (New England Biolabs, Ipswich, MA) was used. Electrophoresis was carried out using constant voltage of 100 volts per gel for the stacking gel for approximately 15 min. The voltage was then adjusted to 80 volts per gel for the separating gel for 90 min.

For the immunoblotting of proteins, the gel was removed from the electrophoresis set and soaked in transfer buffer (25mM Tris-base with 20% methanol, pH 8.5). The 0.45 mm nylon membrane (Osmonics, Trevose, PA) and filter paper (BioRad, Hercules, CA) were presoaked in transfer buffer. The separation gel was placed on the nylon membrane and sandwiched between two pieces of filter papers. The transfer was carried out using BIORAD semi-dry transfer system (BioRad, Hercules, CA) at 15 mA for 30 minutes. After the blotting period, the marker lane was cut out from the membrane. The remaining blot was blocked in PBS (Cellgro, Herndon, VA) containing 5% skim milk (BioRad, Hercules, CA) for one hour to saturate the protein binding sites on the nlyon membrane. The blot was then washed thrice with TBST (25 mM Tris•HCl, pH 8.0, 125 mm NaCl, 0.1% Tween 20). Primary antibody (rabbit polyclonal anti-dengue virus, Abcam, Cambridge, MA) with a dilution of 1:1,000 was added to the blot. The blot was incubated for 2 h at 4°C on an orbital shaker. The membrane was then thoroughly washed with TBST (25 mM Tris•HCl, pH 8.0, 125 mm NaCl, 0.1% Tween 20) thrice for 10 min each. After-which, the blot was incubated with secondary antibody goat anti-rabbit conjugated with HRP (Zymed, San Francisco, CA) at dilution of 1:1,000. The incubation was carried at room temperature for 1 hr on an orbital shaker. After the incubation period, the blot was washed extensively with TBST (25 mM Tris•HCl, pH 8.0, 125 mm NaCl, 0.1% Tween 20) for 3 times with 10 min each time. The blot was developed by addition of Immun-Star HRP luminal substrate for 10 mins and the chemiluminescence signal was detected.

Preparation of Samples for Transmission Electron Microscopy.

A DENV-infected or mock-infected cell monolayer from a 75 cm2 flask was first washed thrice with cold PBS (Cellgro, Herndon, VA) and once with 0.2 M sodium cacodylate buffer, pH 7.4. The cell monolayer was fixed with a primary fixative (2.5 % glutaraldehyde and 2.0 % paraformaldehyde) at 4°C for 24 h. Processing of fixed EM samples was performed by Conventional Electron Microscopy Core Facility (Department of Cell Biology, Harvard Medical School). The cell monolayer was rinsed with 0.2 M cacodylate buffer and scraped off the plastic surface of the tissue culture flask with a cell scraper. The cells were then spun at 1,000 ´ g for 10 min and the supernatant was removed. The cell pellet was then post-fixed with secondary fixative (1% osmium tetroxide) for 2 h at 4°C. A few crystals of potassium ferricyanide (Merck, NJ) were added to the secondary fixative to further enhance the contrast of membranous structures. The cell pellet was washed twice with cacodylate buffer before proceeding to dehydration process through a series of concentrations of ethanol as follows: 50% ethanol, 5 minutes; 70% ethanol, 10 minutes; 95% ethanol, 10 minutes; 100% ethanol, 15 minutes, repeated three times.The dehydrated cell pellet was then infiltrated with a 3:1 ratio of absolute ethanol to LVER for 2 hr at 22 °C. This was followed by a 1:1 ratio for another 2 hr and then 1:3 ratio mixtures which were left overnight. On the following day, absolute LVER was used to further infiltrate the specimen (thrice, each for 2 hr). Embedding and polymerization were done at 60 ºC for about 24 hr in dry gelatin capsules. The embedded specimens were then trimmed (Spencer, UK) and sectioned (50-70 nm thickness) using the ultramicrotome (Ultracut E, UK). The ultrathin sections were picked up onto 200 mesh uncoated copper grid (Taabs, UK) and stained with saturated aqueous uranyl acetate for 5 min. Excess stains were washed off using deionized water. This was then followed by a secondary staining with Reynold's lead citrate solution. The sections were allowed to dry on hot plate and viewed under the Tecnai G2 Spirit Biotwin transmission electron microscope (Hillsboro, OR).