Materials
MEM, DMEM, Opti-MEM, and Pluronic-F68 were purchased from Thermo Fisher Scientific (Waltham, MA, USA). Fetal bovine serum (FBS), penicillin G sodium salt, streptomycin solution, and hypoxanthine-aminopterin-thymidine (HAT) were obtained from MilliporeSigma (Burlington, MA, USA). HyClone™ HyCell™ CHO Medium was purchased from GE Healthcare (Piscataway, NJ, USA). 25 kDa Linear polyethyleneimine (LPEI) was obtained from Polysciences (Warrington, Pennsylvania, USA). Quickantibody-Mouse 5W adjuvant (Cat# KX0210041) was obtained from Biodragon Immunotechnologies (Beijing, China). Bovine serum albumin (BSA) was obtained from Biofroxx (Einhausen, Hessen, Germany). Agarose Gel DNA Extraction Kit, RNAiso Plus, and PrimeScript RT Reagent Kit with gDNA Eraser were bought from TaKaRa (Dalian, Liaoning, China). Rabbit anti-PCSK9 antibody (Cat# ab181142) and rabbit anti-LDLR antibody (Cat# ab52818) were obtained from Abcam (Cambridge, UK). Glutamine, TMB substrate, IPTG, rabbit anti-GAPDH antibody (Cat# D110016), HRP-conjugated goat anti-rabbit IgG (Cat# D110058), and Alexa Fluor 488®-conjugated goat anti-rabbit IgG (Cat# D110061) were bought from BBI (Toronto, ON, Canada). LDL labeled with 1, 1’-dioctadecyl - 3, 3, 3’, 3’-tetramethyl-indocarbocyanine perchlorate (DiI-LDL) was obtained from Yiyuan Biotechnologies (Guangzhou, Guangdong, China). Commercial test kits for LDL-C, TC, TG, and HDL-C were obtained from Nanjing Jiancheng Bioengineering Institute (Nanjing, Jiangsu, China).
Bacterial strains and cell lines
Escherichia coli (E. coli) strains DH5α and BL21 (DE3) were used as hosts for plasmid preparation and single-chain variable fragment (scFv) prokaryotic expression, respectively. Chinese hamster ovary (CHO-3E7) cells were obtained from Genscript Biotech (Nanjing, China), cultured in HyClone™ HyCell™ CHO Medium, and used as hosts for IgG1 eukaryotic transient-expression. Mouse myeloma cell line SP2/0 was purchased from American Type Culture Collection (ATCC, Manassas, VA, USA) and cultured in DMEM medium supplemented with penicillin (100 U/ml), streptomycin (100 µg/ml) and 10% (v/v) FBS. Human hepatic HepG2 cells were obtained from China Infrastructure of Cell Line Resources (Beijing, China) and maintained at 37°C, 5% CO2, in MEM supplemented with penicillin (100 U/ml), streptomycin (100 µg/ml) and 10% (v/v) FBS. All cells were cultured in a humidified incubator at 37°C in an atmosphere of 5% CO2 in the air.
Antigen preparation
To produce human PCSK9 (hPCSK9) protein, the coding sequence of hPCSK9 (GenBank accession number: NM_174936.3) fused with a Kozak consensus sequence (GCCGCCACC) (Hernández et al. 2019) at the 5’-end and a 6×His-tag gene at the 3’-end was synthesized by GenScript Biotech (Nanjing, China) and subcloned into the eukaryotic expression vector pTT5 using the Hind III and EcoR I restriction enzyme sites. The yielded recombinant plasmids were further transiently transfected into suspension CHO-3E7 mammalian cells using PEI transfection reagent as described previously (Stuible et al. 2018). On day 7 post-transfection, the supernatant was purified with a Ni2+ Based immobilized metal ion affinity chromatography (Ni-IMAC, GE Healthcare, Piscataway, NJ, USA), followed by SuperdexTM 200 HR 10/300GL size-exclusion chromatography (GE Healthcare) according to the manufacturer's instructions. Protein concentration was determined by using the BCA protein assay kit (Biomiga, San Diego, USA).
BALB/c mice immunization
Purified hPCSK9 protein (20 µg per mouse) was emulsified with an equal volume of Quickantibody-Mouse 5W adjuvant and intramuscularly injected into the hind legs of female BALB/c mice (6-8-wk, Qinglongshan Experimental Animal Breeding Farm, Nanjing, China) on day 1 and day 21. The final boost (50 µg of hPCSK9 protein) was given intraperitoneally on day 35 without adjuvant. Three days after the final booster immunization, orbital blood of mice was collected for antibody titer detection. When the antibody titters attained 1:100000, the cell fusion was conducted.
Cell fusion and hybridoma screening
According to standard procedures(KÖHler 1975, Kim et al. 2014), splenocytes were harvested from the immunized mice and fused with SP2/0mouse myeloma cells at a ratio of 5:1 using 50% (w/v) polyethylene glycol (PEG) as a fusion regent, and the resulting hybridomas were then cultured in 96-well plates in hypoxanthine-aminopterin-thymidine (HAT) selective medium supplemented with 20% (w/v) FBS. Afterward, the positive hybridoma cells were screened by indirect ELISA and subcloned three times by limiting dilution method. The ascites of identified hybridoma were also prepared by injection of 1×106 positive hybridoma cells into the peritoneal cavity of pristine-treated BALB/c mice, and the ascites containing specific mAbs were purified by protein A affinity chromatography (Roche, Mannheim, Germany). The isotype of purified mAb was determined using a mouse monoclonal subtype identification kit (KMI-2, ProteinTech Group, Chicago, IL, USA) according to the manufacturer’s instruction.
Enzyme-linked immunosorbent assay
The hybridoma cells producing antibodies against hPCSK9 were screened by Enzyme-linked immunosorbent assay (ELISA) using 96-well plates coated with hPCSK9 (1 µg/ml) in coating buffer (0.2 M Na2CO3/NaHCO3, pH 9.6) overnight at 4℃. The plates were then blocked with PBS containing 3% (w/v) bovine serum albumin (BSA) for 2 h at 37℃ and incubated with 100 µl of hybridoma supernatants for 2 h at 37℃. Besides, non-competitive phage ELISA with the addition of increasing concentrations of mAb (10-1, 10, 102, 103, 104, 105 ng/ml) was also set up to further measure the affinity constant (Kaff) of selected mAb as described previously (Beatty et al. 1987). After washing three times with 0.1% Tween in PBS (PBST), HRP-conjugated goat anti-mouse IgG antibody was added and incubated for 1 h at 37℃. Finally, the TMB substrate was added and allowed to develop for 15 minutes at room temperature, and the absorbance at 450 nm was measured using a microplate reader (Thermo Scientific, Waltham, MA, USA).
Western blot analysis
Western blot was performed to detect the protein expression levels of LDLR in HepG2 cells or liver tissues as previously described (Gu et al. 2019). Briefly, the cells or tissues were lysed or homogenized in cold RIPA lysis buffer (Solarbio, Beijing, China) containing 1 mM PMSF on ice for 0.5 h. After centrifugation at 12,000 g for 15 min at 4°C, the cell lysates were collected and total protein concentrations were determined using BCA protein assay. Equal amounts of protein from each sample were subjected to 12% (w/v) SDS-PAGE and transferred to 0.22 µm polyvinylidene fluoride (PVDF) membrane (MerckMillipore, Darmstadt, Germany). After blocking with 0.1% (v/v) TBS-Tween 20 (TBST) containing 5% (w/v) nonfat milk for 2 h at room temperature, the membrane was incubated with corresponding primary antibodies against GAPDH (Cat# D110016, 1:1000) or LDLR (Cat# ab52818, 1:1000) at 4°C overnight, followed by incubation with HRP-conjugated goat anti-rabbit IgG (Cat# D110058, 1:5000) at room temperature for 1 h. Protein bands were detected by enhanced chemiluminescence (ECL) solution (Thermo Scientific, Massachusetts, USA) and quantified by ImageJ software (National Institutes of Health, Bethesda, MD, USA).
LDL-C uptake assay
LDL-C uptake assay was conducted as previously described (Ly et al. 2014, Xu et al. 2020) with slight modification. In brief, HepG2 cells were seeded in black 96-well plates at a density of 3×104 cells per well and cultured overnight. Then, cells were pretreated with opti-MEM for 12 h, followed by treatment with 20 µg/ml hPCSK9 protein alone or co-treatment with 50 µg/ml anti-PCSK9 antibodies for 8 h. Thereafter, 20 µg/ml DiI-LDL was added each well and incubated for an additional 4 h. After washing 3 times with PBS in the dark, LDL-C uptake was measured using a multimode microplate reader (Varioskan lux, Thermo scientific) at 520 nm excitation/580 nm emission.
Cloning of VH and VL gene from hybridoma cells
Total RNA was isolated from hybridoma cells secreting monoclonal antibody (mAb) against hPCSK9 by RNAiso reagent and quantified by measuring A260 nm with Thermo NanoDrop 2000 (Thermo Fisher Scientific). Then the first-strand cDNA was amplified by reverse transcription-polymerase chain reaction (RT-PCR) using PrimeScript™ RT reagent Kit with gDNA Eraser (TaKaRa, Dalian, China) according to the manufacturer's instructions, and the variable region genes of the heavy and light chains of selected mAb were respectively amplified by PCR using PrimeSTAR® HS DNA Polymerase (TaKaRa, Dalian, China) and previously published primer pairs (Wang et al. 2000) with minor modification (Table 1).
Table 1
PCR primers for amplification of VH and VL genes
Primer | Sequence (5’-3’) |
VH-F | CCGGAATTCSARGTNMAGCTGSAGSAGTC |
VH-R | CCCAAGCTTATAGACAGATGGGGGTGTCGTTTTGGC |
VL-F | CCGGAATTCGAYATTGTGMTSACMCARWCTMCA |
VL-R | CCCAAGCTTGGATACAGTTGGTGCAGCATC |
Computer modeling of single-chain variable-fragment antibodies
The three-dimensional (3D) structure models of single-chain variable-fragment antibodies (scFvs) were built via homology modeling using the Schrodinger Suite 2009 (Schrödinger, LLC, New York, NY, USA). Gromacs program, a versatile package in Schrodinger software to perform molecular dynamics, was used to optimize the structure of m5E12scFv in silico and make it closer to the conformation in the natural environment Subsequently, the qualities of the constructed models were evaluated by Ramachandran Plot within Discovery Studio software (Accelrys Software Inc., San Diego, CA, USA). Root mean square deviation (RMSD) is a statistic to assess the deviation degree between resulting and target conformations, which here was used to estimate the model deviation of the constructed models. Thus, the RMSD values of the main chain atoms (C-α, C, N, O) in CDRs between the models were also calculated by Molecular Operating Environment (MOE) software (Chemical Computing Group, Montreal, Canada).
Design of humanized scFvs
Two humanized scFvs were designed based on different antibody humanization methods. The first humanized scFv was designed by transferring murine CDRs onto suitable human consensus FR templates with the highest similarity using a traditional approach called CDR grafting (Verhoeyen et al. 1988). In order to maintain murine CDRs conformation, the key residues, including (a) residues with less than 30% surface accessibility (Pedersen et al. 1994) (b) abnormal residues (Foote et al. 1992), (c) “Vernier” residues located at the FR-CDR junction (De Haard et al. 1999), which may change CDRs conformations were screened by Abcheck (https://www.abcheckantibodies.com/) and Schrodinger Suite 2009. Afterward, another humanized scFv was then designed by back-mutation of these key residues to the amino acids of the original murine mAb.
Construction, expression and purification of scFvs
The genes encoding the humanized scFvs were synthesized at Genscript Biotech and subcloned into the prokaryotic expression vector pET-27b (Novagen, Madison, WI, USA) containing the pectate lyase signal peptide (pelB) of Erwinia carotovora (Blanco-Toribio et al. 2015) using primers listed in Table 2. The resulting recombinant plasmids were further transformed into E. coli BL21 (DE3) cells through CaCl2 heat shock method, and the transformed E. coli BL21 (DE3) cells were cultured in 2×YT medium with 50 µg/ml of kanamycin at 37℃. When the OD600 reached 0.6-0.8, the temperature was shifted to 16℃ and 0.2 mM isopropyl-β-galactosidase (IPTG) was added to induce expression for 18 h. The expressed soluble scFvs with C-terminal 6×His-tag were isolated from the periplasm and purified by Ni-NTA affinity chromatography column (GE Healthcare) and SuperdexTM 75 HR 10/300GL size-exclusion (GE Healthcare) successively, according to manufactures’ protocols.
Table 2
PCR primers for amplification of humanized scFvs
Primer | Sequence (5’-3’) |
m-F | CATGCCATGGATGAAGTTCAGCTGGAGCAGTCAG |
m-R | CCCAAGCTTTCAATGGTGATGGTGATGGTGTTTCAGCTCCAGCTTGGTCC |
h-F | CATGCCATGGATCAGGTGCAGCTGGTGCAGTCTG |
bm-F | CATGCCATGGATGAAGTTCAGCTGGTGCAGTCAG |
h-R/bm-R | CCCAAGCTTTCAATGGTGATGGTGATGGTGTTTGATCTCCACCTTGGTCC |
Saturated site-directed mutagenesis of humanized scFv
The residues in HCDR3 and LCDR3 are most likely to dominate the antibody-antigen interaction (Sundberg et al. 2002). To further identify critical residues involved in hPCSK9 binding, alanine scanning mutagenesis was carried out in these two regions. Briefly, residues in HCDR3 and LCDR3 excepting (a) Tyr and Trp which are advantageous for large van der Waals or hydrophobic interactions, (b) Asn and Ser which mainly form hydrogen bonds, (c) glutamine at the 89th and 90th position of the light chain (H et al. 2015), were selected to mutate to alanine. The oligonucleotide primers used were listed in Table S1. The effect of each mutated site on the activity of humanized scFv was determined by measuring the changes in LDL uptake after treating HepG2 cells with purified humanized scFv proteins. Afterwards, the key residues were identified and site-directed saturation mutagenesis was conducted on these residues using the primers listed in Table S2.
Generation of full-length antibodies
To generate full-length antibodies, the VH and VL of humanized scFvs were fused with the constant region of modified human IgG1 heavy chain (HC) and human kappa light chain (LC, Accession number: ABU90709.2) by overlap-extension PCR (OE-PCR) (Lu et al. 2018), respectively. Primers used for OE-PCR were listed in Table 3. The modified human IgG1 constant region contains several mutations (L234A/L235A/N297G), known to eliminate antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cellular cytotoxicity (CDC) effects (Wines et al. 2000, Hessell et al. 2007, Jefferis 2009, Jacobsen et al. 2017). The C-terminal lysine residue in the heavy chain was also deleted to mitigate mAb heterogeneity caused by C-terminal lysine incomplete cleavage (Dick et al. 2008, Liu et al. 2008). The amplified full-length HC and LC genes containing a Kozak consensus sequence (Hernández 2019) followed by a secretion signal peptide sequence (‘MDWTWRFLFVVAAATGVQS’ for HC secretory expression, Accession number: CAA34971.1; ‘MDMRVPAQLLGLLLLWLSGARC’ for LC secretory expression, Accession number: S24320) at the 5’-end were then subcloned into a mammalian expression vector pTT5 at EcoR I/Hind III restriction sites, respectively. The constructed HC and LC expression plasmids (Fig. S7) were co-transfected into CHO-3E7 mammalian cells at a 1:1 ratio (w:w) for transient expression (Stuible 2018), and the supernatants were purified with a protein A column (Roche, Mannheim, Germany).
Table 3
PCR primers for amplification of full-length antibodies
Primer | Sequence (5’-3’) |
H-F0 | CCGGAATTCGCCGCCACCATGGATTGGACCTGGAGATTTCTGTTTGTGGTGGCCGCCGCCACAGGC |
H-F1 | TTCTGTTTGTGGTGGCCGCCGCCACAGGCGTGCAGTCTCAGGTGCAGCTGGTGCAGTCTGG |
H-R1 | CACGGATGGGCCCTTTGTGCTGGCCGAGGAGACGGTGACCAGGGTCC |
H-F2 | GGACCCTGGTCACCGTCTCCTCGGCCAGCACAAAGGGCCCATCCGTG |
H-R2 | CCCAAGCTTTGGATACAGTTGGTGCAGCATCAGCCCGTTTC |
L-F0 | CCGGAATTCGCCGCCACCATGGACATGAGGGTGCCAGCTCAGCTGCTGGGACTGCTGCTGCTGTGGC |
L-F1 | CAGCTGCTGGGACTGCTGCTGCTGTGGCTGTCCGGAGCTAGGTGCGATATTGTGATGACCCAGTCTCC |
L-R1 | CTTGGAGCGGCCACGGTTCTTTTGATCTCCACCTTGGTCC |
L-F2 | GGACCAAGGTGGAGATCAAAAGAACCGTGGCCGCTCCAAG |
L-R2 | CCCAAGCTTTGGATACAGTTGGTGCAGCATCAGCC |
Binding Affinity Measurement
ForteBio Octet QKe (ForteBio, Fremont, CA, USA), a biomacromolecule interaction analysis system was used to measure the affinity constants of antibodies. According to the manufacturer's instruction, hPCSK9 (50 µg/ml) was biotinylated at room temperature for 2h using a biotinylation kit (Genemore, Shanghai, China) and immobilized on the surface of streptavidin biosensors (ForteBio) for 300 s. The antigen-captured biosensors were then dipped into two-fold series dilution of antibodies for 300 s or longer (association phase) and moved to SD buffer (PBS, pH 7.4, 0.02% Tween 20, 0.1% BSA) without antibodies for 600 s (dissociation phase). The concentrations of scFvs were 6000, 3000, 1500, 750, 375, 187.5, 93.75 nM, and mAbs were 800, 400, 200, 100, 50, 25, and 12.5 nM. The kinetic constants including kon, koff, and KD were analyzed by using ForteBio data analysis software ver. 7.1.
Homology modeling and protein contact identification
To explore the specific binding mechanism of the antigen-antibody, the three-dimensional (3D) structure model of the Fab fragment of h5E12-L230G was constructed by SWISS-MODEL Workspace (http://swissmodel.expasy.org/) based on the top-ranked template with known structure, and the stereochemical property was checked through the Ramachandran plot (Bienert et al. 2017, Waterhouse et al. 2018). Subsequently, the constructed models were further refined and docked with a high-resolution 2.3 Å crystal structure of PCSK9 (PDB ID:5OCA) using the BioLuminate module of Schrödinger Software Suite 2009(Schrödinger). Follow this, the key interaction residues between the antibody and PCSK9 were identified using the Pymol software Version 2.3.0 (Schrödinger), and the free binding energy (△Gbinds) of the Fab-PCSK9-complex were evaluated using the Molecular Mechanics/GB Surface Area (MM/GBSA) method as implemented in HawkDock web server (http://cadd.zju.edu.cn/hawkdock/) (Weng et al. 2019).
Studies in mice
Male C57BL/6 mice aged 6-8 weeks were obtained from Qinglongshan Experimental Animal Breeding Farm (Certificate no. SCXK (Su) 2017-0001; Nanjing, China) and maintained on a 12-h light/dark cycle at room temperature with access to food and water ad libitum. Following 1 week of acclimation, mice were randomly split into 8 groups (a normal group, a model group, and six treatment groups, n=6 per group). On day 1, the model group and dosing group were injected 2 ml saline containing 50 µg pTT5-hPCSK9 intravenously in 5-7s to establish hyperlipidemic mouse model (Miao et al. 2001, Suda et al. 2007), while the normal group was just injected with 2 ml saline. On day 7, the dosing groups were administered with 1, 3, and 10 mg/kg of mAbs in 100 µl saline, respectively, while the normal and model groups were administered same-volume saline. Then the mice were fasted for 8 h and euthanized for blood sample collection. Liver tissues were also collected, and dissected into two parts, one was homogenized by RIPA buffer containing 1mM PMSF for western blot analysis, the other part was fixed in 4% (w/v) paraformaldehyde and embedded in paraffin for immunofluorescence analysis.
Immunofluorescence analysis
Immunofluorescence staining was performed to detect LDLR protein levels in mice livers as previously described (Gu 2019, Xu 2020) with minor modification. Briefly, after deparaffinization and hydration, liver tissue sections were pretreated by heating for 20 min in boiling sodium citrate solution (0.01 M, pH 6.0) for antigen retrieval. Thereafter, the tissue sections were blocked with 10% (v/v) goat serum in PBST for 1 h and incubated with rabbit anti-LDLR antibody (Cat# ab52818, 1:100) overnight at 4°C. After washing three times with PBS, the sections were incubated with Alexa Fluor® 488-conjugated goat anti-rabbit IgG (Cat# D110061, 1:200) at 37℃ for 1 h in the dark and the stained sections were mounted with a drop of glycerin. Images were taken under a Zeiss AX10 fluorescence microscopy (Zeiss, Oberkochen, Germany).
Statistical analysis
Data are expressed as the mean ± SEM of multiple experiments. Comparison between groups was performed using one-way analysis of variance followed by a Tukey multiple comparison test with GraphPad Prism V5.0 software. Results were considered significant when P-values<0.05.