Cell culture
The human embryonic kidney epithelial cell line HEK293T, mouse lung cancer cell line LLC, human lung cancer cell line A549, human liver cancer cell line Huh7, human breast cancer cell lines MDA-MB-231, MCF7, and BT-474 were obtained from American Type Culture Collection (Manassas, VA). ATG7 wild type and knockout HeLa cells were kindly provided by Dr. Masaaki Komatsu in Tokyo Metropolitan Institute of Medical Science. All cell lines except for A549 and MDA-MB-231 were maintained in DMEM medium with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (P/S). A549 and MDA-MB-231 cell lines were maintained in RPMI 1640 and MEM medium with 10% FBS and 1% P/S, respectively. For the hypoxic conditions, hypoxic chambers (ASTEC, Fukuoka, Japan) were used to maintain low oxygen tension (1% O2, 5% CO2, and balanced with N2).
Chemicals
All chemicals except for manassantin A, cruentaren A, anti-PD-1 mAb, AR7, and 17-AAG were purchased from Sigma (St. Louis, MO). Manassantin A and cruentaren A were synthesized according to the literatures [26, 27]. Anti-PD-1 mAb were purchased from BioXCell (West Lebanon, NH). AR7 and 17-AAG were from Selleckchem (Houston, TX).
Mice
Animal experiments were performed using C57BL/6 mice (Japan SLC, Shizuoka, Japan) which were handled in strict compliance with the guidelines for care and use of laboratory animals issued (KNU 2017-145) by the Institutional Ethical Animal Care Committee of Kyungpook National University (Daegu, Korea).
Stable knockdown of LAMP-2A
Control short hairpin scramble (shScramble) and shLAMP-2A constructs (GeneCopoeia, Rockville, M) were used for stable knockdown of LAMP-2A in LLC cells. Cells were infected using lentiviruses containing shScramble and shLAMP-2A followed by selection with puromycin (Sigma) for 10 days.
In vivo tumor allograft experiment and immunofluorescence on frozen tissue section
LLC cells were injected subcutaneously into the flanks of 6-week-old C57BL/6 mice. Vehicle or ManA (5 mg/kg) was administered intraperitoneally every other day (Figure 1B, n=12 per group). Vehicle, ManA (2 mg/kg), 17-AAG (25 mg/kg), anti-PD-1 mAb (5 and 15 mg/kg), or their combination was administered intraperitoneally every other day (Figure 5A, n=6 per group; Figure 6B, n=4 per group). LLC cells expressing shLAMP-2A or carrying a control vector were injected subcutaneously into the flanks of 6-week-old C57BL/6 mice. Vehicle or ManA (5 mg/kg) was administered intraperitoneally every other day (Figure 5D, n=5 per group). Tumor growth was measured with a caliper every other day using the formula, volume = length x width2 x 0.5 (mm3). Tumor tissues were removed at the experimental endpoint and fixed with 4% paraformaldehyde (Sigma). Tissues were dehydrated by serially incubating with 10% to 40% sucrose solution, and embedded in tissue freezing medium (Leica Microsystems, Wetzlar, Germany). Frozen blocks were cut into 50 µm sections, blocked with 5% goat serum in PBST (0.03% Triton X-100 in PBS), and then incubated for 3 h at room temperature (RT) with the following primary anti-Ki67 (Santa Cruz, California, Santa Cruz, CA), anti-CD31 (BD Biosciences, San Jose, CA), anti-HIF-1α (BD Transduction Laboratories, San Jose, CA), anti-GAPDH, anti-IκBα (Cell Signaling Technology, Danvers, MA), anti-LAMP-2A, anti-PD-L1, anti-FOXP3, anti-αSMA (Abcam, Cambridge, UK), anti-CD4, anti-CD8a (BioLegend, San Diego, CA). Frozen blocks were washed with PBST and incubated with specific fluorescent secondary antibodies (Thermo Fisher Scientific) for 1 h. Nuclei were staining with DAPI (Vector Laboratories, Burlingama, CA) and mounted with fluorescent mounting medium (Sigma). To detect the hypoxic area in the tumors, Hypoxyprobe-1™ (60 mg/kg, Natural Pharmacia International, Burlington, MA) was intravenously injected 90 min before tissue fixation. Tumors were harvested, sectioned and stained with MAb1antibody (Natural Pharmacia International, Burlington, MA) and a fluorescent secondary reagent.
Western blot assay
Proteins from cellular lysates were resolved on Tris-Glycine gel or NuPAGE™ 4–12% Bis-Tris Protein Gels (Thermo Fisher Scientific) and transferred to nitrocellulose membranes (Whatman, Maidstone, England). Membranes were incubated overnight at 4 °C with primary antibodies specific for proteins as follows: anti-HIF-1α (BD Transduction Laboratories), anti-PCNA, anti-phospho-AKT (S473), anti-AKT, anti-ATG7, anti-LC3B, anti-TFEB, anti-cleaved caspase-3, anti-α-Tubulin, anti-EPS8 (Cell Signaling Technology), anti-Her2, anti-ATP synthase β (Thermo Fisher Scientific), anti-Hsp90α (Enzo Life Sciences, Farmingdale, NY), anti-LAMP1, anti-β-actin (Santa Cruz Biotechnology), anti-LAMP-2A, anti-LAMP-2B, and anti-Hsp90α (Abcam). Then, the membranes were incubated with horseradish peroxidase-conjugated mouse- or rabbit-IgG (GenTex, Irvine, CA) for 1 h at RT and developed by using West Pico Chemiluminescent Substrate (Pierce, Woburn, MA). The proteins were detected by a digital image analyzer (ImageQuant LAS 4000, GE Healthcare, Buckinghamshire, UK), and the protein bands were obtained within linear range without saturation.
Isolation of the lysosome-rich fraction
Isolation of the lysosome-rich fraction from cells was performed using a kit from BioVision (K235-50) according to the manufacture’s protocol. 2 x 107 cells were pelleted by centrifugation at 600 x g for 10 min, added in 500 μl of Lysosome Isolation Buffer, and incubated on ice for 2 min. Cells were homogenized by passing 30 times through a Glass Dounce Homogenizer. The homogenate was transferred to a fresh tube with 500 μl of Lysosome Enrichment Buffer, and the sample was centrifuged at 500 x g for 10 min at 4 ℃. The supernatant was overlayed on the top of a prepared discontinuous density gradient and gradient centrifuged at 145,000 x g for 2 h at 4 ℃. The lysosome-rich fraction band was collected, mixed with two volumes of PBS, and centrifuged for 30 min at 18,000 x g. Isolated lysosomes were suspended in RIPA buffer, and protein concentration was determined.
RNA isolation and RT-PCR
Total RNA was extracted using a RNeasy Mini Kit (Qiagen, Valncia, CA) and reverse transcribed with a PrimeScript™ RT reagent Kit (Takara Bio Inc., Shiga, Japan). Real-time PCR was performed using an ABI PRISM 7300 Sequence Detection System using SYBR-Green PCR master mix (Applied Biosystems, Foster City, CA) with primer pairs targeting HIF-1α forward, ACCCTCTTCGTCGCTTCGG, and reverse, GCCTCTCCTCAGGTGGCTTG; GLUT1 forward, CAACACGGCCTTCACTGTCG, and reverse, ATGCCAGCGAGGCCTATGAG; PFK forward, CATTTATGTGGGTGCCAAAG, and reverse, ATGATGTTGGAGACGCTCAG; LDHA forward, CATCCTGGGCTATTGGACTCT, and reverse, TGTCCCAAAATGCAAGGAACA; HK2 forward, AAGGTAGAAATGGAGCGAGGT, and reverse, CCCGGAAATTTGTTCCTCCAA; LAMP1 forward, GTGTCTGCTGGACGAGAACA, and reverse, ATGAGGACGATGAGGACCAG; LAMP-2A forward, AACTTCCTTGTGCCCATAGC, and reverse, AGCATGATGGTGCTTGAGAC; LAMP-2B forward, AGAGTGTTCGCTGGATGATG, and reverse, TGCCAATTACGTAAGCAATCA; LAMP-2C forward, AAGGGTTCAGCCTTTCAATG, and reverse, ACAATTATAAGGAAGCCCAAGG; EPS8 forward, CAACCCAGAACAAGCCAGAT, and reverse TGCATTGGAAATCATCCTCA. IκBα forward, TCCTGAGCTCCGAGACTTTC, and reverse, CACGTGTGGCCATTGTAGTT; GAPDH forward, AATCCCATCACCATCTTCCA, and reverse TGGACTCCACGACGTACTC; GFAP forward, AGGAAGATTGAGTCGCTGGA, and reverse ACCTCCTCCTCGTGGATCTT; EEF1A1 forward, CTGTATTGGATTGCCACACG, and reverse GCAGCATCACCAGACTTCAA; RAC1 forward, TGGCTAAGGAGATTGGTGCT, and reverse TCGCTTCGTCAAACACTGTC; RICTOR forward CGGTTGTAGGTTGCCAGTTT, and reverse TCTTTCGGGTTTCATTCCAG; AKT1 TCTATGGCGCTGAGATTGTG, and reverse CTTAATGTGCCCGTCCTTGT; CTSA forward TTCTCTCCAGACCCACTGCT, and reverse GTCACGCATTCCAGGTCTTT; PHLPP1 forward ATCCTGGGCTACACCTTCCT, and reverse CCACAACCCCTTACTGCCTA; β-actin forward, GGATGTCCACGTCACACTTC, and reverse, CACTCTTCCAGCCTTCCTTC. Gene expression was normalized to that of the endogenous reference gene, β-actin.
Lentivirus production
HEK293T cells were transfected with pSIN-PAmCherry-KFERQ-NE expression vector (a gift from Shu Leong Ho, Addgene plasmid #102365) and VirapowerTM packaging mix (Thermo Fisher Scientific) using PolyFect Transfection reagent (QIAGEN, Hilden, Germany) according to the manufacturer’s instructions. After 48 h incubation, supernatant containing lentivirus was harvested.
CMA activity assay
Cells transduced with a lentivirus carrying the PA-mCherry-KFERQ-NE were exposed under UV-A lamp (405 nm) for 5 min. After photoactivation of PA-mCherry-KFERQ-NE proteins, cells were treated with vehicle, ManA, cruentaren A, 6-AN, or 17-AAG under hypoxic conditions. Cells were fixed with 4 % paraformaldehyde (Sigma) and nuclei were stained with DAPI (Vector Laboratories). CMA activity was assessed by counting the number of puncta in cells.
Immunocytochemistry
Cells were fixed with 4% paraformaldehyde (Sigma) and permeabilized with 0.1% Triton X-100. After blocking in 5% normal goat serum (Vector Laboratories, Burlingama, CA) for 1 h, cells were incubated with primary anti-LAMP-2A, anti-HIF-1α (BD Transduction Laboratories), anti-ATP synthase β (Thermo Fisher Scientific), anti-Hsp90α (Enzo Life Sciences), or anti-Flag (Sigma) overnight at 4 °C. Cells were washed with PBS and incubated with specific fluorescent secondary antibodies (Thermo Fisher Scientific) for 1 h. Nuclei were stained with DAPI (Vector Laboratories) and mounted with fluorescent mounting medium (Sigma). Colocalized area was quantified in four images using Image J.
Transfection of siRNA and expression vectors
Cells were transfected with scrambled siRNA, siLAMP-2A, siVPS4A, siVPS4B, siTFEB (Bioneer, Daejeon, South Korea), HIF-1α P402A/P564A double mutant expression vector [28], Myr-p110α expression vector, TFEB-3 X Flag expression vector (kindly provided from Dr. Andrea Ballabio, Baylor College of Medicine), pcDNA3-FLAG-tagged wild-type hHsp90a expression vector (kindly provide from Dr. Leonard M. Neckers, National Cancer Institute), or a control vector using Lipofectamine RNAiMAX reagent (Thermo Fisher Scientific) and TransIT-LT1 Transfection reagent (Mirus Bio, Madison, WI) according to the manufacturer’s instructions
Immunoprecipitation
HEK293T cells were lysed with protein lysis buffer (iNtRON, Daejeon, Korea) containing protease inhibitors (Sigma), and the lysates were incubated with anti-HIF-1α (BD Transduction Laboratories) or anti-ATP synthase β (Thermo Fisher Scientific) at 4 °C for overnight. Slurry protein A/G agarose beads (Pierce, Woburn, MA) were added to reaction mixture, and the mixture was incubated for 3 h at 4 °C. For confirmation of HIF-1α-Hsc70 interaction, cell samples were lysed in lysis buffer (50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 10 mM sodium molybdate, 0.1% NP-40) containing protease inhibitors (Sigma). The lysates were incubated with anti-HIF-1α (BD Transduction Laboratories)-conjugated protein G plus agarose beads (Santa Cruz Biotechnology) for 3 h at 4 °C. Following immunoprecipitation, the beads were washed four times with lysis buffer. Samples were denatured by adding sample buffer in beads and boiling for 10 min. Co-immunoprecipitated proteins were detected by western blot analysis.
Measurement of ATP
The intracellular ATP concentration was measured using ATP Assay Kit (Abcam) according to the manufacturer’s instructions.
Cell proliferation assay
Cells were maintained in the absence or presence of ManA at indicated concentration in complete media under hypoxic conditions for 10 days. Media were changed every 2 days. Cells were fixed and stained with 0.5% crystal violet (Sigma). The quantitative image analyses were performed using ImageJ software.
Measurement of oxygen consumption rate
Oxygen consumption rate (OCR) was measured in a XF24 Extracellular Flux Analyzer (Seahorse Bioscience, North Billerica, MA, USA). Vehicle, ManA (1 μM), oligomycin (1 μM), CCCP (5 μM), rotenone (1 μM), or antimycin A (5 μM) was added at the indicated time points during OCR measurement. The OCR was normalized to the cell number and expressed as a ratio relative to baseline.
Gene expression profiling
MDA-MB-231 cells were treated with vehicle, ManA, geldanamycin, or 17-AAG under hypoxia or normoxia for 24 h. Total RNA was extracted using a mirVana extraction kit (Thermo Fisher Scientific). RNA quality was assessed by electrophoresis. RNA samples were prepared and labelled for interrogation with Affymetrix GeneChip U133 plus 2.0 arrays (Thermo Fisher Scientific) to measure the expression of more than 54,000 probe sets and at least 47,000 transcripts and variants. The raw array data was normalized by RMA for further analysis.
Luciferase refolding assay
ManA and 17-AAG at indicated concentrations in DMSO were added to wells of a white, round-bottom 96-well plate containing 50 μL of cell culture media. Luciferase (pGL4.50-Luc; Promega, Madison, WI)-expressing HEK293T cells were incubated for 10 min at 50 °C in pre-warmed cell culture media until bioluminescence of luciferase was reduced to 1% of the initial counts. Cells were added to wells, and the plate was returned to the incubator for 1 h. After 1 h, 100 μL of luciferase substrate reagent (Promega; 75 mM tricine at pH 7.8, 24 mM MgSO4, 0.3 mM EDTA, 2 mM DTT, 0.313 mM D-luciferin, 0.64 mM coenzyme A, 0.66 mM ATP, 150 mM KCl, 10% Triton X-100, 20% glycerol, and 3.5% DMSO) was added to wells, and the bioluminescence was immediately measured (0.5 s integration time). Cells that were incubated in DMSO were used as 100% bioluminescence (i.e., DMSO = 100% refolding), and the relative refolding for each compound concentration was compared to that in DMSO.
HS-10 resin binding assay
Pig mammary proteins were extracted as previously reported[29]. Gamma-phosphate linked ATP Sepharose slurry was mixed with pig mammary lysate (v/v) for 1 h at RT. Following three washes with buffer A (50mM Tris-HCl pH 7.4, 1 mM DTT, 60 mM MgCl2, 60 mM KCl, 1 µM microcystin) plus 1 M NaCl, the sepharose was mixed with increasing concentrations of HS-10, manassantin A, or ATP (100 mM), and the eluates were mixed with SDS sample buffer and heated for 5 minutes at 90 ºC. Before binding to the HS-10 resin, each sample of lysate was pre-incubated with HS-10 or manassantin A for 1 h at RT. Following the pre-treatment, all samples were tumbled with 100 µL of 50% slurry of resin. After 1 h of tumbling, the resin was washed 3 with a low stringency wash buffer (50 mM Tris-HCl, 60 mM MgCl2, 60 mM KCl, 10 mM citrate, 1 mM DTT, pH 7.4). After the final wash, the buffer was completely aspirated, replaced with SDS sample buffer, and processed as above. Protein gel electrophoresis and silver staining were performed as previously reported[29].
NMR spectroscopy
Hsp90 constructs were prepared according to the previous report [30, 31]. Backbone resonance assignments for the NTD (1-235) and MD (Middle domain, 293-554) proteins were obtained from the previously published data[30, 31]. For NMR titration experiments, 0.1 mM 15N-labeled Hsp90 NTD and Hsp90 MD were prepared. Both the Hsp90 NTD and Hsp90 MD were titrated with addition of two molar equivalents of mannassantin A in the presence of 10% DMSO. The DMSO itself did not disrupt the 3D structure of the proteins since each resonance was well separated in the spectra as the spectrum in the buffer. We could not find any sign of protein aggregation in the 10% DMSO solution. The chemical shift changes of 1H and 15N resonance were measured in a series of 2D [1H,15N]- TROSY spectra, which were recorded at 298 K using a Bruker AVANCE III 600 spectrometer equipped with a cryogenic probe. All NMR spectra were processed using the program NMRPipe[32] and were analyzed with the program NM View[33].
SPR assay
The GST-CTD (C-terminal domain, 464-732) protein of Hsp90 was prepared. Briefly describing, proteins were expressed in LB medium in the E. coli host BL21 with induction at 37 °C for 4–6 h. The GST fused C-terminal domain was purified using the GST-affinity column, GSTPrep FF16/60 (GE Healthcare, Chicago, IL) followed by anion exchange chromatography, HiTrap Q HP and size exclusion chromatography, Sephacryl S100HR column. SPR assay was performed in the Biacore X100 instrument (GE Healthcare). The Anti-GST antibody (40 μl, 0.6 mg/ml) was immobilized on the Biacore sensor chip CM5 (GE Healthcare) using GST-capture kit (GE Healthcare). Activation of the CM5 chip was achieved EDC/NHS solution using a flow rate of 5~10 μl/min. The immobilization of the Anti-GST antibody gave the response units (RU) of 6,000–7,000. The minor fraction of high affinity sites of the polyclonal anti-GST antibody was blocked with recombinant GST (95 μl, 20 μg/ml) at a flow rate of 20 μl/min before capturing of the GST-CTD protein on the antibody. The reference surface should be prepared in the same way as the active surface. The capturing of GST-CTD (55 μl, 100 μM) on the active cell was achieved by the manufacturer’s guide using the capturing protocol that is implemented in the instrument. The surface regeneration of the immobilized chip was achieved with 40 μl of 10 mM glycine-HCl (pH 2.2) injection at a flow rate of 20 μl/min across all flow cells. A serial dilution of manassantin A at 0, 5, 10, 20, 50, and 100.0 μM in 1 × HBS-EP+ buffer (GE Healthcare) supplemented with 2% DMSO were injected over the GST-CTD captured and reference surfaces at contact time, 180 s and flow rate, 10 μl/min, 25°C. To correct the solvent effect originated from DMSO, the step of solvent correction was included during binding experiments. Eight DMSO solutions ranged from 1.5% to 2.8% were prepared and were run at the stage of solvent correction cycle in the assay procedure. The equilibrium dissociation constant (Kd) of manassantin A to the Hsp90 C-terminal domain were analyzed by Evaluation software implemented in the Biacore X100 instrument using the fit models: 1:1 Binding kinetic and Steady State Affinity.
Flow cytometry
To facilitate analysis of lymphoid compartments, tumors were digested with 50 mg/ml collagenase D (Roche) and 10 mg/ml DNase (Roche) per tumor in RPMI 1640 for 1.5 h at 37oC. After pelleting the cells, they were incubated for 5 minutes with ACK lysing buffer (Lonza, Walkersville, MD) and then washed and resuspended in Hank’s balance salt solution (HBSS) (Lonza, Walkersville, MD). Single-cell suspensions of tumors were filtered through a 40 μM nylon strainer (Falcon) and blocked for 10 min on ice with CD16/CD32 Fc block (clone 93) (eBioscience). To detect tumor infiltrated T cell subtypes whole single-cell suspensions were stained with the cell surface markers, PE-cyanine7 CD3e (Thermo Fisher Scientific), PerCP CD8a, (BD Transduction Laboratories), and FITC-CD4 (Invitrogen) antibodies in a FACS buffer, and then analyzed by flow cytometer (FACS Canto II; BD Biosciences). Tumor samples were stained with 1 mg/ml DAPI to exclude dead cells and the flow cytometric analyses were performed using a BD LSR II flow cytometer with FACSDivaTM software.
TUNEL assay
Tumor sections were stained using DeadEndTM Fluormetric TUNEL System (Promega) according to the manufacturer’s instructions.
Patient survival data
The correlation between gene expression and patient outcome has been identified in each cancer type using Prediction of Clinical Outcomes from Genomic Profiles (Precog)[34]. To determine the correlation between patient overall survival and gene expression, gene expression datasets of GEO11969 (only datasets with adenocarcinoma and squamous cell carcinoma patients were included) and TCGA PanCancer Atlas were downloaded from the Gene Expression Omnibus (GEO) repository and cbioportal for lung adenocarcinoma and invasive breast carcinoma. Gene expression of HIF1A, TFEB, and HSP90AA1, or combined expression of TFEB/HIF1A and HSP90AA1/HIF1A was compared with overall patient survival. To acquire statistical significance, gene expression was divided based on median expression value, while quartile cut points were used for single gene expression of TCGA datasets (smallest 25% for TFEB as a negative prognostic factor, top 25% of HSP90AA1 as a positive prognostic factor). Kaplan-Meier curves were plotted using GrapPad Prism 8.4.2 and p values were determined by Log-rank test.
Pharmacokinetics of ManA
C57BL/6 mice were administered ManA at doses of 2 mg/kg (iv; intravenous injection) or 5 mg/kg (po; per oral administration) and venous blood samples were collected at 0.5, 1, 2, 4, 8, and 24 h post-dose. A 20 µL of plasma was separated by centrifugation and stored at -80 °C until analysis. A 20 µL aliquot of plasma samples was deproteinized by addition of 100 µL cold methanol (containing berberine 0.1 ng/mL as internal standard) and centrifuged at 13,200 rpm for 10 min. A 10 µL aliquot of the supernatant was injected into Agilent 6470 LC-MS/MS system. Separation was performed on a Synergy Polar RP column (4 µm particle size, 2.0 mm x 150 mm; Phenomenex, Torrance, CA, USA) using a mobile phase that consisted of methanol and water (85:15, v/v) with 0.1 % formic acid at a flow rate of 0.2 ml/min. Quantification was carried out using multiple reaction monitoring (MRM) at m/z 750.4 → 259.2 for ManA and m/z 336.0 → 320.0 for berberin (internal standard) in positive ionization mode and collision energy of 25 eV.
The pharmacokinetic parameters were determined by the non-compartmental analysis (WinNonlin® 5.1; Pharsight, Mountain View, CA, USA). The maximum plasma concentration (Cmax) and time to reach Cmax (Tmax) values were obtained from the plasma concentration-time curves. The area under the plasma concentration-time curve from zero to infinity (AUC) was calculated by the trapezoidal-extrapolation method. Oral bioavailability (BA) was calculated as follows: BA (%) = AUCp.o. normalized by po dose/AUCi.v. normalized by iv dose × 100.
Plasma and metabolic stability of Man A
ManA stock solution (10 mM dissolved in methanol) was diluted with potassium phosphate buffer (pH7.4) to make a concentration of 1 µM. A 10 µL aliquot of this solution was then added to 80 µL aliquot of 100 mM potassium phosphate buffer (pH7.4) containing human or mouse liver microsomes (80 µL of 0.625 mg protein/ml phosphate buffer) and incubated at 37 °C for 10 min before adding of 10 µL of an NADPH regenerating system to start the reaction. Reactions were stopped at 0, 15, 30, 45, and 60 min time-points by the addition of 200 µL cold methanol (containing 0.1 ng/mL berberine as internal standard) and vortexed for 15 min and centrifuged at 16,000 × g for 5 min. A 5 µL aliquot of the supernatant was injected into the LC-MS/MS system. Positive control study using 1 μM metformin (for high microsomal stability) and 1 μM propranolol (for low microsomal stability) was also performed to ensure the system feasibility. For plasma stability, 100 μL of mouse or human plasma containing 1 μM of each compound (Man A, metformin, or propranolol) was incubated for 2 h at 37 °C on a shaking incubator at 300 rpm. Reactions were stopped at 0, 30, 60, 90, and 120 min time-points by the addition of 200 µL cold methanol (containing 0.1 ng/mL berberine as internal standard). Thereafter, the mixture was vortexed for 15 min and centrifuged at 16,000 × g for 5 min. After centrifugation, a 5 μL aliquot was injected into the LC–MS/MS system. The ratio of peak area of test compound /internal standard was used to determine the % remaining of each compound over time. The half- life and % compound remaining after 60 min were then calculated.
Caco-2 permeability
Caco-2 cells (passage no 42-43; purchased from ATCC, Rockville, MD, USA) were seeded on collagen-coated 12-transwell membranes at a density of 5 × 105 cells/mL and maintained at 37 °C in a humidified atmosphere with 5% CO2/95% air for 21 d. The culture medium was replaced every 2 d and the permeability assay was conducted as previously described. Briefly, HBSS medium containing marker compounds (caffeine, propranolol, ofloxacin, atenolol, and paclitaxel) (2, 5, 10, 50 µM, and 20 µM, respectively) or ManA (20 µM) was added to the donor side and aliquots (100 μL) from the receiver side were collected at 15, 30, 45, and 60 min and stored at -80 °C until further analysis. The thawed samples were added to 300 μL methanol (containing berberine as internal standard) and centrifuged at 13,200 rpm for 10 min. A 5 µl aliquot of the supernatant was injected into the LC-MS/MS system.
For the permeability calculation, the transport rate of ginsenosides and marker compounds was calculated from the slope of the regression line from the mean permeated amounts vs. incubation time plot. The apparent permeability (Papp) was calculated from the following equation:
Statistical analysis
All data were expressed as mean ± standard deviations (SD) from at least 3 samples. Statistical comparisons were analyzed using the Student’s t-test. Differences with p < 0.05 and p < 0.01 were considered statistically significant and highly significant, respectively.