Please wait a minute...
Protein & Cell

ISSN 1674-800X

ISSN 1674-8018(Online)

CN 11-5886/Q

Postal Subscription Code 80-984

2018 Impact Factor: 7.575

Prot Cell    2011, Vol. 2 Issue (7) : 573-584    https://doi.org/10.1007/s13238-011-1076-4      PMID: 21822802
RESEARCH ARTICLE
Pharmacological applications of a novel neoepitope antibody to a modified amyloid precursor protein-derived beta-secretase product
Guoxin Wu1(), Sethu Sankaranarayanan1, Donna L. Montgomery2, Adam J. Simon1, Zhiqiang An2, Mary J. Savage1
1. Department of Neurology, Merck Research Laboratory, West Point, PA 19486, USA; 2. Department of Biologics Research, Merck Research Laboratory, West Point, PA 19486, USA
 Download: PDF(520 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

We have previously described a novel artificial NFEV β-secretase (BACE1) cleavage site, which when introduced into the amyloid-β precursor protein (APP), significantly enhances APP cleavage by BACE1 in in vitro and cellular assays. In this study, we describe the identification and characterization of a single chain fragment of variable region (scFv), specific to the EV neo-epitope derived from BACE1 cleavage of the NFEV-containing peptide, and its conversion to IgG1. Both the scFv displayed on phage and EV-IgG1 show exquisite specificity for binding to the EV neoepitope without cross-reactivity to other NFEV containing peptides or WT-APP KMDA cleavage products. EV-IgG1 can detect as little as 0.3 nmol/L of the EV peptide. EV-IgG1 antibody was purified, conjugated with alkaline phosphatase and utilized in various biological assays. In the BACE1 enzymatic assay using NFEV substrate, a BACE1 inhibitor MRK-3 inhibited cleavage with an IC50 of 2.4 nmol/L with excellent reproducibility. In an APP_NFEV stable SH-SY5Y cellular assay, the EC50 for inhibition of EV-Aβ peptide secretion with MRK-3 was 236 nmol/L, consistent with values derived using an EV polyclonal antibody. In an APP_NFEV knock-in mouse model, both Aβ_EV40 and Aβ_EV42 peptides in brain homogenate showed excellent gene dosage dependence. In conclusion, the EV neoepitope specific monoclonal antibody is a novel reagent for BACE1 inhibitor discovery for both in vitro, cellular screening assays and in vivo biochemical studies. The methods described herein are generally applicable to novel synthetic substrates and enzyme targets to enable robust screening platforms for enzyme inhibitors.

Keywords scFv      antibody      BACE1      amyloid-β precursor protein (APP)      immunoassay     
Corresponding Author(s): Wu Guoxin,Email:Guoxin_wu@merck.com   
Issue Date: 01 July 2011
 Cite this article:   
Guoxin Wu,Sethu Sankaranarayanan,Donna L. Montgomery, et al. Pharmacological applications of a novel neoepitope antibody to a modified amyloid precursor protein-derived beta-secretase product[J]. Prot Cell, 2011, 2(7): 573-584.
 URL:  
https://academic.hep.com.cn/pac/EN/10.1007/s13238-011-1076-4
https://academic.hep.com.cn/pac/EN/Y2011/V2/I7/573
Peptide namePeptide amino acid sequence
NFEV substrateSEVNFEVEFRHDSGYK-biotin
EV cleavage productEVEFRHDSGYK-biotin
CSPa 1 with NFBiotin-KTEEISEVNF
CSP 2 with NBiotin-KTEEISEVN
CSP 3 with FEVFEVEFRHDSGYK-biotin
CSP 4 with NFEBiotin-KTEEISEVNFE
CSP 5 with VVEFRHDSGYK-biotin
CSP 6 with KMBiotin-KTEEISEVKM
CSP 7 with DADAEFRHDSGYK-biotin
CSP 8 with NLBiotin-KTEEISEVNL
Tab.1  Design of biotin labeled peptides
Fig.1  
Eight scFv clones (EV1–EV8) were identified that bind specifically to the EV peptide but not to the NFEV peptide.
Fig.1  
Eight scFv clones (EV1–EV8) were identified that bind specifically to the EV peptide but not to the NFEV peptide.
Fig.2  
Amino acid differences in complementary determining regions (CDR) 1, 2 and 3 of both heavy and light chains are bold and underlined. CDR portions in both heavy and light chains among the 8 clones are aligned and underlined. The amino acid sequence in CDR region shows that clone EV2 is the same as EV6 and EV3 is identical to EV7.
Fig.2  
Amino acid differences in complementary determining regions (CDR) 1, 2 and 3 of both heavy and light chains are bold and underlined. CDR portions in both heavy and light chains among the 8 clones are aligned and underlined. The amino acid sequence in CDR region shows that clone EV2 is the same as EV6 and EV3 is identical to EV7.
Fig.3  
The human APP_WT BACE cleavage site is indicated by KMDA, while the Swedish mutation is NLDA and the artificial BACE substrate is the NFEV sequence. The secretase cleavage sites are indicated by arrows. α, alpha-secretase; β, beta-secretase; γ, gamma-secretase. Cleavage of the APP_NFEV sequence by β-secretase followed by γ-secretase leads to the production of Aβ_EV40 and Aβ_EV42 peptides.
Fig.3  
The human APP_WT BACE cleavage site is indicated by KMDA, while the Swedish mutation is NLDA and the artificial BACE substrate is the NFEV sequence. The secretase cleavage sites are indicated by arrows. α, alpha-secretase; β, beta-secretase; γ, gamma-secretase. Cleavage of the APP_NFEV sequence by β-secretase followed by γ-secretase leads to the production of Aβ_EV40 and Aβ_EV42 peptides.
Fig.4  
The identified EV-specific phage clones were counter-screened using an array of peptides (Table 1) to assess their specificity for binding to the N-terminal EV peptide. All 5 identified EV reactive clones (EV1, EV2, EV3, EV5 and EV8) were highly specific to the N-terminal EV peptide.
Fig.4  
The identified EV-specific phage clones were counter-screened using an array of peptides (Table 1) to assess their specificity for binding to the N-terminal EV peptide. All 5 identified EV reactive clones (EV1, EV2, EV3, EV5 and EV8) were highly specific to the N-terminal EV peptide.
Fig.5  
(A) EV3-IgG1 shows robust signal with the EV N-terminal peptide and greater than 6000-fold signal to noise ratio without detectable cross reactivity to other N or C-terminal modified peptides. (B) The EV rabbit polyclonal antibody showed significant reactivity with other counter-screening peptides in addition to the EV N-terminal peptide. (C) Sensitivity for the detection of synthetic EV N-terminal peptide by EV3-IgG-AP. The lower limit of reliable quantitation of the EV peptide is ~0.3 nmol/L.
Fig.5  
(A) EV3-IgG1 shows robust signal with the EV N-terminal peptide and greater than 6000-fold signal to noise ratio without detectable cross reactivity to other N or C-terminal modified peptides. (B) The EV rabbit polyclonal antibody showed significant reactivity with other counter-screening peptides in addition to the EV N-terminal peptide. (C) Sensitivity for the detection of synthetic EV N-terminal peptide by EV3-IgG-AP. The lower limit of reliable quantitation of the EV peptide is ~0.3 nmol/L.
Fig.6  
(A) BACE1 concentration response in an BACE1 enzymatic reaction using NFEV substrate, followed by detection of the EV N-terminal peptide with EV3-IgG1-AP. A linear dose-dependent increase in EV peptide signal is observed as a function of BACE1 concentration ( = 0.99, <0.001, = 3). (B) Time course of production of EV N-terminal peptides following cleavage of NFEV substrate by 0.3 nmol/L BACE1 in an enzymatic reaction. A linear increase in EV N-terminal peptide signal was observed over time ( = 0.912, <0.001, = 3). (C) A specific BACE1 inhibitor MRK-3 was tested at a range of concentrations in the BACE1 enzymatic reaction with NFEV peptide substrate. A dose-dependent inhibition of EV N-terminal peptide production was observed with MRK-3 with an IC of 2.4 nmol/L.
Fig.6  
(A) BACE1 concentration response in an BACE1 enzymatic reaction using NFEV substrate, followed by detection of the EV N-terminal peptide with EV3-IgG1-AP. A linear dose-dependent increase in EV peptide signal is observed as a function of BACE1 concentration ( = 0.99, <0.001, = 3). (B) Time course of production of EV N-terminal peptides following cleavage of NFEV substrate by 0.3 nmol/L BACE1 in an enzymatic reaction. A linear increase in EV N-terminal peptide signal was observed over time ( = 0.912, <0.001, = 3). (C) A specific BACE1 inhibitor MRK-3 was tested at a range of concentrations in the BACE1 enzymatic reaction with NFEV peptide substrate. A dose-dependent inhibition of EV N-terminal peptide production was observed with MRK-3 with an IC of 2.4 nmol/L.
Intra-dayInter-day
Mean (nmol/L)2.112.14
SD (nmol/L)0.260.24
Min1.801.81
Max2.292.40
% CV12.3911.17
Tab.2  IC for BACE1 inhibitor MRK-3 using NFEV substrate ( = 5)
Fig.7  
(A) Production of EV_Aβ peptides from SH-SY5Y cells stably expressing APP_NFEV was tested at a range of concentrations of MRK-3. EV_Aβ peptides were measured using the detecting antibody EV3-IgG1-AP in a sandwich ELISA following capture with 4G8. A dose-dependent inhibition of EV-Aβ production was observed with an EC of ~236 nmol/L. (B) Measurement of EV_Aβ peptides using an EV rabbit polyclonal detecting antibody yielded comparable EC of ~254 nmol/L in the same experiment as in (A). (C) MRK-3 treatment led to a dose-dependent increase in sAPPα (EC ~332 nmol/L) from the same experiment as in (A), indicating no cellular toxicity. (D) Assay specificity in EV_Aβ detection with EV3-IgG1-AP antibody. Immunodepletion of Aβ peptides with Aβ-selective antibodies 6E10 or 4G8, but not mouse IgG control antibody led to a complete loss of signal in the EV3-IgG1-AP detection system.
Fig.7  
(A) Production of EV_Aβ peptides from SH-SY5Y cells stably expressing APP_NFEV was tested at a range of concentrations of MRK-3. EV_Aβ peptides were measured using the detecting antibody EV3-IgG1-AP in a sandwich ELISA following capture with 4G8. A dose-dependent inhibition of EV-Aβ production was observed with an EC of ~236 nmol/L. (B) Measurement of EV_Aβ peptides using an EV rabbit polyclonal detecting antibody yielded comparable EC of ~254 nmol/L in the same experiment as in (A). (C) MRK-3 treatment led to a dose-dependent increase in sAPPα (EC ~332 nmol/L) from the same experiment as in (A), indicating no cellular toxicity. (D) Assay specificity in EV_Aβ detection with EV3-IgG1-AP antibody. Immunodepletion of Aβ peptides with Aβ-selective antibodies 6E10 or 4G8, but not mouse IgG control antibody led to a complete loss of signal in the EV3-IgG1-AP detection system.
Fig.8  
Brain Aβ gene-dosage dependence, with homozygote (Homo) displaying 2 fold higher levels than heterozygote (Heter) litter mates. Wild type animals show no detectable signal. (A) Aβ_EV40 and (B) Aβ_EV42 levels. ( = 3, Mean±SEM).
Fig.8  
Brain Aβ gene-dosage dependence, with homozygote (Homo) displaying 2 fold higher levels than heterozygote (Heter) litter mates. Wild type animals show no detectable signal. (A) Aβ_EV40 and (B) Aβ_EV42 levels. ( = 3, Mean±SEM).
1 An, Z., Forrest, G., Moore, R., Cukan, M., Hayto, P., Huang, L., Vitelli, S., Zhao, J., Lu, P., Hua, J., Gibson, C.R., Harvey, B.R., Montgomery, D., Zaller, D., Wang, F., Strohl, W. (2009). IgG2m4, an engineered antibody isotype with reduced Fc function. mAbs 1 , 572–579 .
2 Cai, H., Wang, Y., McCarthy, D., Wen, H., Borchelt, D.R., Price, D.L., and Wong, P.C. (2001). BACE1 is the major β-secretase for generation of Abeta peptides by neurons.Nat Neurosci 4, 233–234 .
pmid:11224536
3 Copeland, R.A. (2005). Evaluation of enzyme inhibitors in drug discovery. A guide for medicinal chemists and pharmacologists.Methods Biochem Anal 46, 1–265 .
pmid:16350889
4 Harrison, S.M., Harper, A.J., Hawkins, J., Duddy, G., Grau, E., Pugh, P.L., Winter, P.H., Shilliam, C.S., Hughes, Z.A., Dawson, L.A., (2003). BACE1 (beta-secretase) transgenic and knockout mice: identification of neurochemical deficits and behavioral changes.Mol Cell Neurosci 24, 646–655 .
pmid:14664815
5 McCampbell, A., Wessner, K., Marlatt, M.W., Wolffe, C., Toolan, D., Podtelezhnikov, A., Yeh, S., Zhang, R., Szczerba, P., Tanis, K.Q., (2011). Induction of Alzheimer’s-like changes in brain of mice expressing mutant APP fed excess methionine.J Neurochem 116, 82–92 .
pmid:21054384
6 Miller, M.D., Geleziunas, R., Bianchi, E., Lennard, S., Hrin, R., Zhang, H., Lu, M., An, Z., Ingallinella, P., Finotto, M., (2005). A human monoclonal antibody neutralizes diverse HIV-1 isolates by binding a critical gp41 epitope.Proc Natl Acad Sci U S A 102, 14759–14764 .
pmid:16203977
7 Montgomery, D.L., Wang, Y., Hrin, R., Luftig, M., Su, B., Miller, M.D., Wang, F., Haytko, P., Huang, L., Vitelli, S., Condra, J., Liu, X., Hampton, R., Carfi, A., Pessi, A., Bianchi, E., Joyce, J., Lloyd, C., Geleziunas, R., Bramhill, D., King, V.M., Finnefrock, A.C., Strohl, W., An, Z. (2009). Affinity maturation and characterization of a human monoclonal antibody against HIV-1 gp41. mAbs 1 , 462–474 .
8 Palys, T.J., Schmid, K.E., Scherer, J.M., and Schoepp, R.J. (2006). Conversion of a mouse Fab into a whole humanized IgG antibody for detecting botulinum toxin.Hum Antibodies 15, 125–132 .
pmid:17522434
9 Pietrak, B.L., Crouthamel, M.C., Tugusheva, K., Lineberger, J.E., Xu, M., DiMuzio, J.M., Steele, T., Espeseth, A.S., Stachel, S.J., Coburn, C.A., (2005). Biochemical and cell-based assays for characterization of BACE-1 inhibitors.Anal Biochem 342, 144–151 .
pmid:15958191
10 Rüfenacht, P., Güntert, A., Bohrmann, B., Ducret, A., and D?beli, H. (2005). Quantification of the A beta peptide in Alzheimer’s plaques by laser dissection microscopy combined with mass spectrometry.J Mass Spectrom 40, 193–201 .
pmid:15706631
11 Sankaranarayanan, S., Holahan, M.A., Colussi, D., Crouthamel, M.C., Devanarayan, V., Ellis, J., Espeseth, A., Gates, A.T., Graham, S.L., Gregro, A.R., (2009). First demonstration of cerebrospinal fluid and plasma A beta lowering with oral administration of a beta-site amyloid precursor protein-cleaving enzyme 1 inhibitor in nonhuman primates.J Pharmacol Exp Ther 328, 131–140 .
pmid:18854490
12 Sankaranarayanan, S., Price, E.A., Wu, G., Crouthamel, M.C., Shi, X.P., Tugusheva, K., Tyler, K.X., Kahana, J., Ellis, J., Jin, L., (2008). In vivo beta-secretase 1 inhibition leads to brain Abeta lowering and increased alpha-secretase processing of amyloid precursor protein without effect on neuregulin-1.J Pharmacol Exp Ther 324, 957–969 .
pmid:18156464
13 Shi, X., Tugusheva, K., Bruce, J.E., Luca, A., Chen, D.E., Hu, B., Wu, G.X., Price, E., Register, R.B., Lineberger, J., (2005). Novel mutation introduced at the β-site of amyloid precursor protein enhances β-secretase cleavage in vitro and in cells.J Alzheimers Dis 7, 139–148
pmid:15851852
14 Shi, X.P., Chen, E., Yin, K.C., Na, S., Garsky, V.M., Lai, M.T., Li, Y.M., Platchek, M., Register, R.B., Sardana, M.K., (2001). The pro domain of β-secretase does not confer strict zymogen-like properties but does assist proper folding of the protease domain.J Biol Chem 276, 10366–10373 .
pmid:11266439
15 Simon, A.J., Chen, L., Price, E., Xu, M., Lucka, A., Tang, M., Chen, E., Espeseth, A.S., Sardana, M., Shi, X.P., (2004). A genetically engineered mouse model with an enhanced beta-secretase substrate exhibits increased amyloid generation.Neurobiol Aging 25, S242.
16 Sinha, S., and Lieberburg, I. (1999). Cellular mechanisms of β-amyloid production and secretion.Proc Natl Acad Sci U S A 96, 11049–11053 .
pmid:10500121
17 Stachel, S.J., Coburn, C.A., Steele, T.G., Jones, K.G., Loutzenhiser, E.F., Gregro, A.R., Rajapakse, H.A., Lai, M.T., Crouthamel, M.C., Xu, M., (2004). Structure-based design of potent and selective cell-permeable inhibitors of human beta-secretase (BACE-1).J Med Chem 47, 6447–6450 .
pmid:15588077
18 Tomasselli, A.G., Qahwash, I., Emmons, T.L., Lu, Y., Leone, J.W., Lull, J.M., Fok, K.F., Bannow, C.A., Smith, C.W., Bienkowski, M.J., (2003). Employing a superior BACE1 cleavage sequence to probe cellular APP processing.J Neurochem 84, 1006–1017 .
pmid:12603825
19 Turner, R.T. 3rd, Koelsch, G., Hong, L., Castanheira, P., Ermolieff, J., Ghosh, A.K., and Tang, J. (2001). Subsite specificity of memapsin 2 (β-secretase): implications for inhibitor design.Biochemistry 40, 10001–10006 .
pmid:11513577
20 Vassar, R., and Citron, M. (2000). Abeta-generating enzymes: recent advances in β- and γ-secretase research.Neuron 27, 419–422 .
pmid:11055423
21 Vaughan, T.J., Williams, A.J., Pritchard, K., Osbourn, J.K., Pope, A.R., Earnshaw, J.C., McCafferty, J., Hodits, R.A., Wilton, J., and Johnson, K.S. (1996). Human antibodies with sub-nanomolar affinities isolated from a large non-immunized phage display library.Nat Biotechnol 14, 309–314 .
pmid:9630891
22 Willem, M., Garratt, A.N., Novak, B., Citron, M., Kaufmann, S., Rittger, A., DeStrooper, B., Saftig, P., Birchmeier, C., and Haass, C. (2006). Control of peripheral nerve myelination by the beta-secretase BACE1.Science 314, 664–666 .
pmid:16990514
23 Wu, G., Sankaranarayanan, S., Hsieh, S.H., Simon, A.J., and Savage, M.J. (2011). Decrease in brain soluble amyloid precursor protein β (sAPPβ) in Alzheimer’s disease cortex.J Neurosci Res 89, 822–832 .
pmid:21433051
24 Wu, G., Sankaranarayanan, S., Tugusheva, K., Kahana, J., Seabrook, G., Shi, X.P., King, E., Devanarayan, V., Cook, J.J., and Simon, A.J. (2008). Decrease in age-adjusted cerebrospinal fluid β-secretase activity in Alzheimer’s subjects.Clin Biochem 41, 986–996 .
pmid:18489907
25 Yang, L.B., Lindholm, K., Yan, R., Citron, M., Xia, W., Yang, X.L., Beach, T., Sue, L., Wong, P., Price, D., (2003). Elevated β-secretase expression and enzymatic activity detected in sporadic Alzheimer disease.Nat Med 9, 3–4 .
pmid:12514700
[1] Xinhua Wang, Mary Mathieu, Randall J. Brezski. IgG Fc engineering to modulate antibody effector functions[J]. Protein Cell, 2018, 9(1): 63-73.
[2] Liming Liu. Pharmacokinetics of monoclonal antibodies and Fc-fusion proteins[J]. Protein Cell, 2018, 9(1): 15-32.
[3] Kyoji Tsuchikama, Zhiqiang An. Antibody-drug conjugates: recent advances in conjugation and linker chemistries[J]. Protein Cell, 2018, 9(1): 33-46.
[4] William R. Strohl. Current progress in innovative engineered antibodies[J]. Protein Cell, 2018, 9(1): 86-120.
[5] Xin Wang, Zhiqiang An, Wenxin Luo, Ningshao Xia, Qinjian Zhao. Molecular and functional analysis of monoclonal antibodies in support of biologics development[J]. Protein Cell, 2018, 9(1): 74-85.
[6] Rudo Simeon, Zhilei Chen. In vitro-engineered non-antibody protein therapeutics[J]. Protein Cell, 2018, 9(1): 3-14.
[7] Xuyuan Zhang, Pan Yang, Nan Wang, Jialong Zhang, Jingyun Li, Hao Guo, Xiangyun Yin, Zihe Rao, Xiangxi Wang, Liguo Zhang. The binding of a monoclonal antibody to the apical region of SCARB2 blocks EV71 infection[J]. Protein Cell, 2017, 8(8): 590-600.
[8] Shuguang Tan,Danqing Chen,Kefang Liu,Mengnan He,Hao Song,Yi Shi,Jun Liu,Catherine W.-H. Zhang,Jianxun Qi,Jinghua Yan,Shan Gao,George F. Gao. Crystal clear: visualizing the intervention mechanism of the PD-1/PD-L1 interaction by two cancer therapeutic monoclonal antibodies[J]. Protein Cell, 2016, 7(12): 866-877.
[9] Haiyan Chu,Ting Wu,Wenyu Wu,Wenzhen Tu,Shuai Jiang,Sidi Chen,Yanyun Ma,Qingmei Liu,Xiaodong Zhou,Li Jin,Jiucun Wang. Involvement of collagen-binding heat shock protein 47 in scleroderma-associated fibrosis[J]. Protein Cell, 2015, 6(8): 589-598.
[10] Yuanxi Kang, Jia Guo, Zhiwei Chen. Closing the door to human immunodeficiency virus[J]. Prot Cell, 2013, 4(2): 86-102.
[11] Tengteng Zhang, Yun Xia, Lijuan Zhang, Wanrong Bao, Chao Hong, Xiao-Ming Gao. CD1dhiCD5+ B cells differentiate into antibody-secreting cells under the stimulation with calreticulin fragment[J]. Prot Cell, 2013, 4(11): 872-881.
[12] Shaopeng Chen, Junkang Qiu, Chuan Chen, Chunchun Liu, Yuheng Liu, Lili An, Junying Jia, Jie Tang, Lijun Wu, Haiying Hang. Affinity maturation of anti-TNF-alpha scFv with somatic hypermutation in non-B cells[J]. Prot Cell, 2012, 3(6): 460-469.
[13] Meng Xu, Xuexiang Du, Mingyue Liu, Sirui Li, Xiaozhu Li, Yang-Xin Fu, Shengdian Wang. The tumor immunosuppressive microenvironment impairs the therapy of anti-HER2/neu antibody[J]. Prot Cell, 2012, 3(6): 441-449.
[14] Hui Dai, Xiao-Ming Gao. Elevated levels of serum antibodies against alpha-1, 6-glucan in patients with systemic lupus erythematosus or rheumatoid arthritis[J]. Prot Cell, 2011, 2(9): 739-744.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed