Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2022, Vol. 16 Issue (4) : 584-595    https://doi.org/10.1007/s11684-021-0844-7
RESEARCH ARTICLE
Long-term correction of hemorrhagic diathesis in hemophilia A mice by an AAV-delivered hybrid FVIII composed of the human heavy chain and the rat light chain
Jianhua Mao1(), Yun Wang1,2, Wei Zhang1, Yan Shen3, Guowei Zhang4, Wenda Xi5, Qiang Wang1, Zheng Ruan1, Jin Wang2, Xiaodong Xi1()
1. Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
2. Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics and Department of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
3. Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
4. The School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
5. Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
 Download: PDF(1603 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Conventional therapies for hemophilia A (HA) are prophylactic or on-demand intravenous FVIII infusions. However, they are expensive and inconvenient to perform. Thus, better strategies for HA treatment must be developed. In this study, a recombinant FVIII cDNA encoding a human/rat hybrid FVIII with an enhanced procoagulant potential for adeno-associated virus (AAV)-delivered gene therapy was developed. Plasmids containing human FVIII heavy chain (hHC), human light chain (hLC), and rat light chain (rLC) were transfected into cells and hydrodynamically injected into HA mice. Purified AAV viruses were intravenously injected into HA mice at two doses. Results showed that the hHC+ rLC protein had a higher activity than the hHC+ hLC protein at comparable expression levels. The specific activity of hHC+ rLC was about 4- to 8-fold higher than that of their counterparts. Hydrodynamic injection experiments obtained consistent results. Notably, the HA mice undergoing the AAV-delivered hHC+ rLC treatment exhibited a visibly higher activity than those treated with hHC+ hLC, and the therapeutic effects lasted for up to 40 weeks. In conclusion, the application of the hybrid FVIII (hHC+ rLC) via an AAV-delivered gene therapy substantially improved the hemorrhagic diathesis of the HA mice. These data might be of help to the development of optimized FVIII expression cassette for HA gene therapy.

Keywords hemophilia A      adeno-associated virus (AAV)      human/rat hybrid factor VIII      gene therapy      dual chain strategy     
Corresponding Author(s): Jianhua Mao,Xiaodong Xi   
About author:

Tongcan Cui and Yizhe Hou contributed equally to this work.

Just Accepted Date: 19 August 2021   Online First Date: 14 January 2022    Issue Date: 02 September 2022
 Cite this article:   
Jianhua Mao,Yun Wang,Wei Zhang, et al. Long-term correction of hemorrhagic diathesis in hemophilia A mice by an AAV-delivered hybrid FVIII composed of the human heavy chain and the rat light chain[J]. Front. Med., 2022, 16(4): 584-595.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-021-0844-7
https://academic.hep.com.cn/fmd/EN/Y2022/V16/I4/584
Fig.1  Activity and antigen expression levels of hHC+ hLC and hHC+ rLC in transfected 293T cells. (A) FVIII activity of hHC+ hLC and hHC+ rLC. Expression plasmid of hHC driven by CB promoter was transfected together with hLC or rLC into 293T cells at a ratio of 1:1 (HC:LC), and the total amount of the plasmids was 1 mg/well. At 48 h after transfection, the medium was collected, and FVIII activity was detected via aPTT assay. Normal human plasma (NHP) was used as the standard, and initial activity was defined as one unit. (B) Detection of FVIII antigen by ELISA. FVIII antigen was measured with a specific antibody to hHC. NHP was used as the standard, and one unit was calculated as 0.2 mg. (C) Specific activity of FVIII calculated on the basis of ELISA. The specific activity of FVIII was calculated on the basis of the data from A and B. (D) Detection of FVIII expression by WB. Expression of FVIII in reduced and nonreduced states was detected by WB. The samples used were the transfected cell media of hHC+ hLC and hHC+ rLC. The intensity ratio of hHC+ hLC and hHC+ rLC is shown at the bottom. The intensity ratio of hHC+ hLC was calibrated as 1.0, and the relative intensity of hHC+ rLC to hHC+ hLC was 1.1. (E) The specific activity of FVIII calculated on the basis of WB. The specific activity of FVIII was calculated on the basis of the data from A and D. (n = 3, ***P<0.001.)
Fig.2  Activity and antigen expression of hHC+ hLC and hHC+ rLC in transfected HepG2 cells. (A) Detection of FVIII activity. The expression plasmids of hHC, which were driven by the ApoE-hAAT promoter, were co-transfected into HepG2 cells with LC (hLC and rLC). The total amount of plasmids of each group was 1 mg, and the ratio of HC:LC was 1:1. The activity of FVIII of each group was detected at 48 h post-transfection via aPTT assay. Normal human plasma (NHP) was used as the standard, and initial activity was defined as one unit. (B) Detection of FVIII antigen by ELISA. The FVIII antigens of hHC+ hLC and hHC+ rLC were monitored by ELISA, and the samples used were the same as those utilized in aPTT assay. NHP was used as the standard, and one unit was calculated as 0.2 mg. (C) Detection of FVIII expression by WB. Expression of FVIII of hHC+ hLC and hHC+ rLC in reduced and nonreduced states was detected by WB. The intensity ratio of hHC+ hLC and hHC+ rLC is shown at the bottom. The intensity ratio of hHC+ hLC was calibrated as 1.0, and the relative intensity of hHC+ rLC to hHC+ hLC was 1.03. (D) Specific activity of FVIII calculated on the basis of ELISA. Specific activity of FVIII was calculated on the basis of the data from A and B. (E) Specific activity of FVIII calculated on the basis of WB. Specific activity of FVIII was calculated on the basis of the data from A and D. (n = 3, ***P<0.001.)
Fig.3  Detection of the stability of hHC+ hLC and hHC+ rLC in 293T and HepG2 cells. (A) The sample was the culture medium from the transfected 293T cells. It was incubated at 37?°C for 2, 6, 12, and 24 h. (B) The sample was the culture medium from the transfected HepG2 cells. It was incubated at 37?°C for 2, 4, 6, 8, 10, 12, and 24 h. At the indicated time points, the residual FVIII activities of the samples were measured by aPTT (n = 3). Relative activity was calculated according to the activity before incubation at 37?°C, which was defined as 1.0 AU (arbitrary unit).
Fig.4  Detection of FVIII activity, specific activity, TEG, and hemoglobin concentration in the hydrodynamically injected HA mice. (A) Detection of FVIII activity. Expression plasmids for hHC were hydrodynamically injected into HA mice together with hLC and rLC at a ratio of 1:1. A total of 150 mg plasmid diluted in 2 mL saline was injected into each HA mouse through the lateral tail vein. Plasma was collected at 48 h after injection. FVIII activity was detected by aPTT (*P<0.05, **P<0.01, ***P<0.001). Normal human plasma (NHP) was used as the standard. (B) The FVIII antigen was detected using hHC antibody and ELISA. NHP was employed as the standard. (C) Specific FVIII activity was calculated on the basis of the data from A and B (****P<0.0001). (D) Detection of TEG. Fresh citrated blood sample was harvested from the hydrodynamically injected HA mice at 54 h after injection and immediately used for TEG assay. (E) Detection of hemoglobin concentration. Citrated peripheral blood (eye-bleeding) was collected after 6 h from the mouse whose tail was half-clipped. Hb concentration was measured using an animal blood cell counter.
Fig.5  Detection of the activity and specific activity of hHC+ hLC and hHC+ rLC in AAV-mediated gene therapy in HA mice in vivo. (A) Detection of FVIII activity in the intermediate dose group. AAV viruses delivering hHC together with hLC and rLC at a ratio of 1:1 were injected into HA mice through the lateral tail vein at the intermediate dose group (8×1012 vg/kg), and FVIII activity was measured using aPTT assays from 0 to 40 weeks (*P<0.05). Normal human plasma (NHP) was used as the standard. (B) Detection of FVIII activity in the high dose group (2×1013 vg/kg). The ratio and the injection method were the same as those in the intermediate dose group. FVIII activity was measured from 0 to 40 weeks (*P<0.05, ***P<0.001). (C) The FVIII antigen of the mice from the high dose group was detected using ELISA at 16 and 24 weeks. hHC antibody was used to detect FVIII expression, and NHP was employed as the standard. (D) Specific FVIII activity at 16 and 24 weeks was calculated on the basis of the data from B and C (*P<0.05).
Fig.6  Detection of liver function. (A) ALT was measured in HA mice administered with a high dose of hHC+ hLC and hHC+ rLC (n = 4). (B) AST was measured in HA mice administered with a high dose of hHC+ hLC and hHC+ rLC (n = 4). The reference range of ALT or AST is displayed at the top of the bar chart. The data were from the corresponding HA mice before AAV-delivered FVIII was administered.
1 M Makris, J Oldenburg, EP Mauser-Bunschoten, K Peerlinck, G Castaman, K; subcommittee on Factor VIII, Factor IX and Rare Bleeding Disorders. FijnvandraatThe definition, diagnosis and management of mild hemophilia A: communication from the SSC of the ISTH. J Thromb Haemost 2018; 16(12): 2530–2533
https://doi.org/10.1111/jth.14315 pmid: 30430726
2 PHB Bolton-Maggs, KJ Pasi. Haemophilias A and B. Lancet 2003; 361(9371): 1801–1809
https://doi.org/10.1016/S0140-6736(03)13405-8 pmid: 12781551
3 J Lai, C Hough, J Tarrant, D Lillicrap. Biological considerations of plasma-derived and recombinant factor VIII immunogenicity. Blood 2017; 129(24): 3147–3154
https://doi.org/10.1182/blood-2016-11-750885 pmid: 28432221
4 M Morfini, CAP Rapisarda. Safety of recombinant coagulation factors in treating hemophilia. Expert Opin Drug Saf 2019; 18(2): 75–85
https://doi.org/10.1080/14740338.2019.1574743 pmid: 30681006
5 GQ Perrin, RW Herzog, DM Markusic. Update on clinical gene therapy for hemophilia. Blood 2019; 133(5): 407–414
https://doi.org/10.1182/blood-2018-07-820720 pmid: 30559260
6 C Borsotti, A Follenzi. New technologies in gene therapy for inducing immune tolerance in hemophilia A. Expert Rev Clin Immunol 2018; 14(12): 1013–1019
https://doi.org/10.1080/1744666X.2018.1539667 pmid: 30345839
7 A Asokan, DV Schaffer, RJ Samulski. The AAV vector toolkit: poised at the clinical crossroads. Mol Ther 2012; 20(4): 699–708
https://doi.org/10.1038/mt.2011.287 pmid: 22273577
8 AK Zaiss, Q Liu, GP Bowen, NC Wong, JS Bartlett, DA Muruve. Differential activation of innate immune responses by adenovirus and adeno-associated virus vectors. J Virol 2002; 76(9): 4580–4590
https://doi.org/10.1128/JVI.76.9.4580-4590.2002 pmid: 11932423
9 AC Nathwani, EGD Tuddenham, S Rangarajan, C Rosales, J McIntosh, DC Linch, P Chowdary, A Riddell, AJ Pie, C Harrington, J O’Beirne, K Smith, J Pasi, B Glader, P Rustagi, CYC Ng, MA Kay, J Zhou, Y Spence, CL Morton, J Allay, J Coleman, S Sleep, JM Cunningham, D Srivastava, E Basner-Tschakarjan, F Mingozzi, KA High, JT Gray, UM Reiss, AW Nienhuis, AM Davidoff. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med 2011; 365(25): 2357–2365
https://doi.org/10.1056/NEJMoa1108046 pmid: 22149959
10 S Rangarajan, L Walsh, W Lester, D Perry, B Madan, M Laffan, H Yu, C Vettermann, GF Pierce, WY Wong, KJ Pasi. AAV5-Factor VIII gene transfer in severe hemophilia A. N Engl J Med 2017; 377(26): 2519–2530
https://doi.org/10.1056/NEJMoa1708483 pmid: 29224506
11 AC Nathwani, AM Davidoff, EGD Tuddenham. Gene therapy for hemophilia. Hematol Oncol Clin North Am 2017; 31(5): 853–868
https://doi.org/10.1016/j.hoc.2017.06.011 pmid: 28895852
12 AC Nathwani, AW Nienhuis, AM Davidoff. Current status of gene therapy for hemophilia. Curr Hematol Rep 2003; 2(4): 319–327
pmid: 12901329
13 B Nambiar, C C Sookdeo, P Berthelette, R Jackson, S Piraino, B Burnham, S Nass, D Souza, CR O’Riordan, KA Vincent, SH Cheng, D Armentano, S Kyostio-Moore. Characteristics of minimally oversized adeno-associated virus vectors encoding human factor VIII generated using producer cell lines and triple transfection. Hum Gene Ther Methods 2017; 28(1): 23–38
https://doi.org/10.1089/hgtb.2016.124 pmid: 28166648
14 C Mah, R Sarkar, I Zolotukhin, M Schleissing, X Xiao, HH Kazazian, BJ Byrne. Dual vectors expressing murine factor VIII result in sustained correction of hemophilia A mice. Hum Gene Ther 2003; 14(2): 143–152
https://doi.org/10.1089/104303403321070838 pmid: 12614565
15 Q Wang, B Dong, J Firrman, S Roberts, AR Moore, W Cao, Y Diao, P Kapranov, R Xu, W Xiao. Efficient production of dual recombinant adeno-associated viral vectors for factor VIII delivery. Hum Gene Ther Methods 2014; 25(4): 261–268
https://doi.org/10.1089/hgtb.2014.093 pmid: 25093498
16 BS Doshi, VR Arruda. Gene therapy for hemophilia: what does the future hold? Ther Adv Hematol 2018; 9(9): 273–293
https://doi.org/10.1177/2040620718791933 pmid: 30210756
17 SA Shestopal, JJ Hao, E Karnaukhova, Y Liang, MV Ovanesov, M Lin, JH Kurasawa, TK Lee, JH Mcvey, AG Sarafanov. Expression and characterization of a codon-optimized blood coagulation factor VIII. J Thromb Haemost 2017; 15(4): 709–720
https://doi.org/10.1111/jth.13632 pmid: 28109042
18 J McIntosh, PJ Lenting, C Rosales, D Lee, S Rabbanian, D Raj, N Patel, EGD Tuddenham, OD Christophe, JH McVey, S Waddington, AW Nienhuis, JT Gray, P Fagone, F Mingozzi, SZ Zhou, KA High, M Cancio, CYC Ng, J Zhou, CL Morton, AM Davidoff, AC Nathwani. Therapeutic levels of FVIII following a single peripheral vein administration of rAAV vector encoding a novel human factor VIII variant. Blood 2013; 121(17): 3335–3344
https://doi.org/10.1182/blood-2012-10-462200 pmid: 23426947
19 GN Nguyen, LA George, JI Siner, RJ Davidson, CB Zander, XL Zheng, VR Arruda, RM Camire, DE Sabatino. Novel factor VIII variants with a modified furin cleavage site improve the efficacy of gene therapy for hemophilia A. J Thromb Haemost 2017; 15(1): 110–121
https://doi.org/10.1111/jth.13543 pmid: 27749002
20 MP Kosloski, KA Shetty, H Wakabayashi, PJ Fay, SV Balu-Iyer. Effects of replacement of factor VIII amino acids Asp519 and Glu665 with Val on plasma survival and efficacy in vivo. AAPS J 2014; 16(5): 1038–1045
https://doi.org/10.1208/s12248-014-9627-2 pmid: 24934295
21 CB Doering, JF Healey, ET Parker, RT Barrow, P Lollar. High level expression of recombinant porcine coagulation factor VIII. J Biol Chem 2002; 277(41): 38345–38349
https://doi.org/10.1074/jbc.M206959200 pmid: 12138172
22 CB Doering, JF Healey, ET Parker, RT Barrow, P Lollar. Identification of porcine coagulation factor VIII domains responsible for high level expression via enhanced secretion. J Biol Chem 2004; 279(8): 6546–6552
https://doi.org/10.1074/jbc.M312451200 pmid: 14660593
23 Q Wang, B Dong, J Firrman, W Wu, S Roberts, AR Moore, LS Liu, MPS Chin, Y Diao, J Kost, W Xiao. Evaluation of the biological differences of canine and human factor VIII in gene delivery: implications in human hemophilia treatment. Gene Ther 2016; 23(7): 597–605
https://doi.org/10.1038/gt.2016.34 pmid: 27064790
24 DE Sabatino, CF Freguia, R Toso, A Santos, EP Merricks, HH Kazazian, TC Nichols, RM Camire, VR Arruda. Recombinant canine B-domain-deleted FVIII exhibits high specific activity and is safe in the canine hemophilia A model. Blood 2009; 114(20): 4562–4565
https://doi.org/10.1182/blood-2009-05-220327 pmid: 19770361
25 P Lollar, ET Parker, PJ Fay. Coagulant properties of hybrid human/porcine factor VIII molecules. J Biol Chem 1992; 267(33): 23652–23657
https://doi.org/10.1016/S0021-9258(18)35888-5 pmid: 1429706
26 W Zhang, J Mao, Y Shen, G Zhang, Y Shao, Z Ruan, Y Wang, W Wu, X Wang, J Zhu, S Chen, W Xiao, X Xi. Evaluation of the activity levels of rat FVIII and human FVIII delivered by adeno-associated viral vectors both in vitro and in vivo. Blood Cells Mol Dis 2018; 73: 47–54
https://doi.org/10.1016/j.bcmd.2018.09.004 pmid: 30249384
27 CD Scallan, T Liu, AE Parker, SL Patarroyo-White, H Chen, H Jiang, J Vargas, D Nagy, SK Powell, JF Wright, R Sarkar, HH Kazazian, A McClelland, LB Couto. Phenotypic correction of a mouse model of hemophilia A using AAV2 vectors encoding the heavy and light chains of FVIII. Blood 2003; 102(12): 3919–3926
https://doi.org/10.1182/blood-2003-01-0222 pmid: 12893764
28 C Mueller, D Ratner, L Zhong, M Esteves-Sena, G Gao. Production and discovery of novel recombinant adeno-associated viral vectors. Curr Protoc Microbiol 2012; Chapter 14: s26
29 J Wang, J Xie, H Lu, L Chen, B Hauck, RJ Samulski, W Xiao. Existence of transient functional double-stranded DNA intermediates during recombinant AAV transduction. Proc Natl Acad Sci USA 2007; 104(32): 13104–13109
https://doi.org/10.1073/pnas.0702778104 pmid: 17664425
30 Y Kuang, J Wang, X Lu, S Lu, L Zhang, C Shen, J Fei, Z Wang. Generation of factor VIII gene knockout mouse by tetraploid embryo complementation technology. Chin J Med Genet (Zhonghua Yi Xue Yi Chuan Xue Za Zhi) 2010; 27(1): 1–6 (in Chinese)
pmid: 20140858
31 D Wang, G Zhang, J Gu, X Shao, Y Dai, J Li, X Pan, S Yao, A Xu, Y Jin, J Huang, Q Shi, J Zhu, X Xi, Z Chen, S Chen. In vivo generated hematopoietic stem cells from genome edited induced pluripotent stem cells are functional in platelet-targeted gene therapy of murine hemophilia A. Haematologica 2020; 105(4): e175–e179
https://doi.org/10.3324/haematol.2019.219089 pmid: 31296582
32 A Tiede. Half-life extended factor VIII for the treatment of hemophilia A. J Thromb Haemost 2015; 13(Suppl 1): S176–S179
https://doi.org/10.1111/jth.12929 pmid: 26149020
33 E Persson, T Foscolo, M Hansen. Reagent-specific underestimation of turoctocog alfa pegol (N8-GP) clotting activity owing to decelerated activation by thrombin. Res Pract Thromb Haemost 2019; 3(1): 114–120
https://doi.org/10.1002/rth2.12167 pmid: 30656284
34 AD Shapiro, MV Ragni, R Kulkarni, J Oldenberg, A Srivastava, DV Quon, KJ Pasi, H Hanabusa, I Pabinger, J Mahlangu, P Fogarty, D Lillicrap, S Kulke, J Potts, S Neelakantan, I Nestorov, S Li, JA Dumont, H Jiang, A Brennan, GF Pierce. Recombinant factor VIII Fc fusion protein: extended-interval dosing maintains low bleeding rates and correlates with von Willebrand factor levels. J Thromb Haemost 2014; 12(11): 1788–1800
https://doi.org/10.1111/jth.12723 pmid: 25196897
35 G Lippi, EJ Favaloro. Emicizumab (ACE910): clinical background and laboratory assessment of hemophilia A. Adv Clin Chem 2019; 88: 151–167
https://doi.org/10.1016/bs.acc.2018.10.003 pmid: 30612605
36 KA High, XM Anguela. Adeno-associated viral vectors for the treatment of hemophilia. Hum Mol Genet 2016; 25(R1): R36–R41
https://doi.org/10.1093/hmg/ddv475 pmid: 26614390
37 L Lisowski, AP Dane, K Chu, Y Zhang, SC Cunningham, EM Wilson, S Nygaard, M Grompe, IE Alexander, MA Kay. Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature 2014; 506(7488): 382–386
https://doi.org/10.1038/nature12875 pmid: 24390344
38 LA George, SK Sullivan, A Giermasz, JEJ Rasko, BJ Samelson-Jones, J Ducore, A Cuker, LM Sullivan, S Majumdar, J Teitel, CE McGuinn, MV Ragni, AY Luk, D Hui, JF Wright, Y Chen, Y Liu, K Wachtel, A Winters, S Tiefenbacher, VR Arruda, JCM van der Loo, O Zelenaia, D Takefman, ME Carr, LB Couto, XM Anguela, KA High. Hemophilia B gene therapy with a high-specific-activity factor IX variant. N Engl J Med 2017; 377(23): 2215–2227
https://doi.org/10.1056/NEJMoa1708538 pmid: 29211678
39 T Okuyama, RM Huber, W Bowling, R Pearline, SC Kennedy, MW Flye, KP Ponder. Liver-directed gene therapy: a retroviral vector with a complete LTR and the ApoE enhancer-α1-antitrypsin promoter dramatically increases expression of human α1-antitrypsin in vivo. Hum Gene Ther 1996; 7(5): 637–645
https://doi.org/10.1089/hum.1996.7.5-637 pmid: 8845389
40 P Simioni, D Tormene, G Tognin, S Gavasso, C Bulato, NP Iacobelli, JD Finn, L Spiezia, C Radu, VR Arruda. X-linked thrombophilia with a mutant factor IX (factor IX Padua). N Engl J Med 2009; 361(17): 1671–1675
https://doi.org/10.1056/NEJMoa0904377 pmid: 19846852
41 M Takeyama, H Wakabayashi, PJ Fay. Contribution of factor VIII light-chain residues 2007–2016 to an activated protein C-interactive site. Thromb Haemost 2013; 109(2): 187–198
https://doi.org/10.1160/TH12-08-0561 pmid: 23224054
42 LM O’Brien, M Mastri, PJ Fay. Regulation of factor VIIIa by human activated protein C and protein S: inactivation of cofactor in the intrinsic factor Xase. Blood 2000; 95(5): 1714–1720
https://doi.org/10.1182/blood.V95.5.1714.005k40_1714_1720 pmid: 10688829
43 KP Pratt, BW Shen, K Takeshima, EW Davie, K Fujikawa, BL Stoddard. Structure of the C2 domain of human factor VIII at 1.5 A resolution. Nature 1999; 402(6760): 439–442
https://doi.org/10.1038/46601 pmid: 10586887
44 VA Novakovic, DB Cullinan, H Wakabayashi, PJ Fay, JD Baleja, GE Gilbert. Membrane-binding properties of the Factor VIII C2 domain. Biochem J 2011; 435(1): 187–196
https://doi.org/10.1042/BJ20101797 pmid: 21210768
45 M Watzka, C Geisen, E Seifried, J Oldenburg. Sequence of the rat factor VIII cDNA. Thromb Haemost 2004; 91(1): 38–42
https://doi.org/10.1160/TH03-06-0336 pmid: 14691566
[1] Yanlin Zhao,Xiao Zhong,Xiaohong Ou,Huawei Cai,Xiaoai Wu,Rui Huang. Cotransfecting norepinephrine transporter and vesicular monoamine transporter 2 genes for increased retention of metaiodobenzylguanidine labeled with iodine 131 in malignant hepatocarcinoma cells[J]. Front. Med., 2017, 11(1): 120-128.
[2] Wei Lu,Qingzhang Zhou,Hao Yang,Hao Wang,Yexing Gu,Qi Shen,Jinglun Xue,Xiaoyan Dong,Jinzhong Chen. Gene therapy for hemophilia B mice with scAAV8-LP1-hFIX[J]. Front. Med., 2016, 10(2): 212-218.
[3] Chuanfeng Wu, Cynthia E. Dunbar. Stem cell gene therapy: the risks of insertional mutagenesis and approaches to minimize genotoxicity[J]. Front Med, 2011, 5(4): 356-371.
[4] Jian XIN BM, Ze-Feng XIA MD, Kai-Xiong TAO MD, Kai-Lin CAI PhD, Gao-Xiong HAN MD, Xiao-Ming SHUAI MD, Ji-Liang WANG MD, Han-Song DU MD, Guo-Bin WANG PhD, Yan LUO MM, . Development of a magnetite-gene complex for gene transfection[J]. Front. Med., 2010, 4(2): 241-246.
[5] Xudong YU MM, Zengwu SHAO MD, Liming XIONG MD, Weiwei XU MM, Hezhong WANG MM, Huifa XU MM, . Adenovirus-mediated tissue inhibitor of metalloproteinase-3 gene transfection inhibits rabbit intervertebral disc degeneration in vivo[J]. Front. Med., 2009, 3(4): 415-420.
[6] Youguo HAO, Min ZHANG, Jinzhi XU, Bitao BU, Jiajun WEI. Construction of lentiviral vector carrying Rab9 gene and its expression in mouse brain[J]. Front Med Chin, 2009, 3(2): 141-147.
[7] XIA Xi, WANG Beibei, CAO Li, CHEN Gang, WU Peng, LU Yunping, ZHOU Jianfeng, MA Ding. Investigation of gene therapy of denovirus in immune suppression[J]. Front. Med., 2008, 2(4): 386-390.
[8] XIONG Ying, GUO Wen, LI Ting, LI Ke. Influence of Survivin-targeted siRNA on the biological features of colorectal carcinoma cells[J]. Front. Med., 2007, 1(3): 304-307.
[9] TIAN Yongji, LI Guilin, GAO Jun, WANG Renzhi, KONG Yanguo, ZHANG Zhenxing, LI Shifang, TIAN Shiqiang, DOU Wanchen, ZHANG Bo. Construction of 6HRE-GFAP-Baxα system specific for glioma gene therapy[J]. Front. Med., 2007, 1(1): 49-53.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed