1
|
England C, TrejoMartinez J, PerezSanchez P, Karki U, Xu J. Plants as Biofactories for Therapeutic Proteins and Antiviral Compounds to Combat COVID-19. Life (Basel) 2023; 13:617. [PMID: 36983772 PMCID: PMC10054913 DOI: 10.3390/life13030617] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) had a profound impact on the world's health and economy. Although the end of the pandemic may come in 2023, it is generally believed that the virus will not be completely eradicated. Most likely, the disease will become an endemicity. The rapid development of vaccines of different types (mRNA, subunit protein, inactivated virus, etc.) and some other antiviral drugs (Remdesivir, Olumiant, Paxlovid, etc.) has provided effectiveness in reducing COVID-19's impact worldwide. However, the circulating SARS-CoV-2 virus has been constantly mutating with the emergence of multiple variants, which makes control of COVID-19 difficult. There is still a pressing need for developing more effective antiviral drugs to fight against the disease. Plants have provided a promising production platform for both bioactive chemical compounds (small molecules) and recombinant therapeutics (big molecules). Plants naturally produce a diverse range of bioactive compounds as secondary metabolites, such as alkaloids, terpenoids/terpenes and polyphenols, which are a rich source of countless antiviral compounds. Plants can also be genetically engineered to produce valuable recombinant therapeutics. This molecular farming in plants has an unprecedented opportunity for developing vaccines, antibodies, and other biologics for pandemic diseases because of its potential advantages, such as low cost, safety, and high production volume. This review summarizes the latest advancements in plant-derived drugs used to combat COVID-19 and discusses the prospects and challenges of the plant-based production platform for antiviral agents.
Collapse
Affiliation(s)
- Corbin England
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
- Molecular Biosciences Program, Arkansas State University, Jonesboro, AR 72401, USA
| | | | - Paula PerezSanchez
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Uddhab Karki
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
- Molecular Biosciences Program, Arkansas State University, Jonesboro, AR 72401, USA
| | - Jianfeng Xu
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA
- College of Agriculture, Arkansas State University, Jonesboro, AR 72401, USA
| |
Collapse
|
2
|
Kim NA, An IB, Lim HS, Yang SI, Jeong SH. Biophysical evaluation of hybrid Fc fusion protein of hGH to achieve basal buffer system. Int J Pharm 2016; 513:421-430. [DOI: 10.1016/j.ijpharm.2016.09.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/05/2016] [Accepted: 09/18/2016] [Indexed: 12/22/2022]
|
3
|
Urbanowicz RA, Lacek K, Lahm A, Bienkowska-Szewczyk K, Ball JK, Nicosia A, Cortese R, Pessi A. Cholesterol conjugation potentiates the antiviral activity of an HIV immunoadhesin. J Pept Sci 2016; 21:743-9. [PMID: 26292842 DOI: 10.1002/psc.2802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 01/04/2023]
Abstract
Immunoadhesins are engineered proteins combining the constant domain (Fc) of an antibody with a ligand-binding (adhesion) domain. They have significant potential as therapeutic agents, because they maintain the favourable pharmacokinetics of antibodies with an expanded repertoire of ligand-binding domains: proteins, peptides, or small molecules. We have recently reported that the addition of a cholesterol group to two HIV antibodies can dramatically improve their antiviral potency. Cholesterol, which can be conjugated at various positions in the antibody, including the constant (Fc) domain, endows the conjugate with affinity for the membrane lipid rafts, thus increasing its concentration at the site where viral entry occurs. Here, we extend this strategy to an HIV immunoadhesin, combining a cholesterol-conjugated Fc domain with the peptide fusion inhibitor C41. The immunoadhesin C41-Fc-chol displayed high affinity for Human Embryonic Kidney (HEK) 293 cells, and when tested on a panel of HIV-1 strains, it was considerably more potent than the unconjugated C41-Fc construct. Potentiation of antiviral activity was comparable to what was previously observed for the cholesterol-conjugated HIV antibodies. Given the key role of cholesterol in lipid raft formation and viral fusion, we expect that the same strategy should be broadly applicable to enveloped viruses, for many of which it is already known the sequence of a peptide fusion inhibitor similar to C41. Moreover, the sequence of heptad repeat-derived fusion inhibitors can often be predicted from genomic information alone, opening a path to immunoadhesins against emerging viruses.
Collapse
Affiliation(s)
- Richard A Urbanowicz
- School of Life Sciences, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom.,Nottingham Digestive Diseases Centre Biomedical Research Unit, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Krzysztof Lacek
- CEINGE, Via Gaetano Salvatore 486, 80145, Napoli, Italy.,Laboratory of Virus Molecular Biology, University of Gdansk, 80-822, Gdansk, Poland
| | - Armin Lahm
- PeptiPharma, Viale Città D'Europa 679, 00144, Roma, Italy
| | | | - Jonathan K Ball
- School of Life Sciences, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom.,Nottingham Digestive Diseases Centre Biomedical Research Unit, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Alfredo Nicosia
- CEINGE, Via Gaetano Salvatore 486, 80145, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Napoli, Italy
| | | | - Antonello Pessi
- CEINGE, Via Gaetano Salvatore 486, 80145, Napoli, Italy.,PeptiPharma, Viale Città D'Europa 679, 00144, Roma, Italy.,JV Bio, Via Gaetano Salvatore 486, 80145, Napoli, Italy
| |
Collapse
|
4
|
Novarra S, Grinberg L, Rickert KW, Barnes A, Wilson S, Baca M. A hingeless Fc fusion system for site-specific cleavage by IdeS. MAbs 2016; 8:1118-25. [PMID: 27210548 DOI: 10.1080/19420862.2016.1186321] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Fusion of proteins to the Fc region of IgG is widely used to express cellular receptors and other extracellular proteins, but cleavage of the fusion partner is sometimes required for downstream applications. Immunoglobulin G-degrading enzyme of Streptococcus pyogenes (IdeS) is a protease with exquisite specificity for human IgG, and it can also cleave Fc-fusion proteins at a single site in the N-terminal region of the CH2 domain. However, the site of IdeS cleavage results in the disulfide-linked hinge region partitioning with the released protein, complicating downstream usage of the cleaved product. To tailor the Fc fragment for release of partner proteins by IdeS treatment, we investigated the effect of deleting regions of IgG-derived sequence that are upstream of the cleavage site. Elimination of the IgG-derived hinge sequence along with several residues of the CH2 domain had negligible effects on expression and purity of the fusion protein, while retaining efficient processing by IdeS. An optimal Fc fragment comprising residues 235-447 of the human IgG1 heavy chain sufficed for efficient production of fusion proteins and minimized the amount of residual Ig-derived sequence on the cleavage product following IdeS treatment. Pairing of this truncated Fc fragment with IdeS cleavage enables highly specific cleavage of Fc-fusion proteins, thus eliminating the need to engineer extraneous cleavage sequences. This system should be helpful for producing Fc-fusion proteins requiring downstream cleavage, particularly those that are sensitive to internal miscleavage if treated with alternative proteases.
Collapse
Affiliation(s)
- Shabazz Novarra
- a Department of Antibody Discovery and Protein Engineering , MedImmune LLC , Gaithersburg , MD , USA
| | - Luba Grinberg
- a Department of Antibody Discovery and Protein Engineering , MedImmune LLC , Gaithersburg , MD , USA
| | - Keith W Rickert
- a Department of Antibody Discovery and Protein Engineering , MedImmune LLC , Gaithersburg , MD , USA
| | - Arnita Barnes
- a Department of Antibody Discovery and Protein Engineering , MedImmune LLC , Gaithersburg , MD , USA
| | - Susan Wilson
- a Department of Antibody Discovery and Protein Engineering , MedImmune LLC , Gaithersburg , MD , USA
| | - Manuel Baca
- a Department of Antibody Discovery and Protein Engineering , MedImmune LLC , Gaithersburg , MD , USA
| |
Collapse
|
5
|
Lim JY, Kim NA, Lim DG, Eun CY, Choi D, Jeong SH. Biophysical stability of hyFc fusion protein with regards to buffers and various excipients. Int J Biol Macromol 2016; 86:622-9. [DOI: 10.1016/j.ijbiomac.2016.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/31/2016] [Accepted: 02/01/2016] [Indexed: 11/25/2022]
|
6
|
Wright GJ, Bianchi E. The challenges involved in elucidating the molecular basis of sperm-egg recognition in mammals and approaches to overcome them. Cell Tissue Res 2015. [PMID: 26224538 PMCID: PMC4700105 DOI: 10.1007/s00441-015-2243-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Sexual reproduction is used by many different organisms to create a new generation of genetically distinct progeny. Cells originating from separate sexes or mating types segregate their genetic material into haploid gametes which must then recognize and fuse with each other in a process known as fertilization to form a diploid zygote. Despite the central importance of fertilization, we know remarkably little about the molecular mechanisms that are involved in how gametes recognize each other, particularly in mammals, although the proteins that are displayed on their surfaces are almost certainly involved. This paucity of knowledge is largely due to both the unique biological properties of mammalian gametes (sperm and egg) which make them experimentally difficult to manipulate, and the technical challenges of identifying interactions between membrane-embedded cell surface receptor proteins. In this review, we will discuss our current knowledge of animal gamete recognition, highlighting where important contributions to our understanding were made, why particular model systems were helpful, and why progress in mammals has been particularly challenging. We discuss how the development of mammalian in vitro fertilization and targeted gene disruption in mice were important technological advances that triggered progress. We argue that approaches employed to discover novel interactions between cell surface gamete recognition proteins should account for the unusual biochemical properties of membrane proteins and the typically highly transient nature of their interactions. Finally, we describe how these principles were applied to identify Juno as the egg receptor for sperm Izumo1, an interaction that is essential for mammalian fertilization.
Collapse
Affiliation(s)
- Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Enrica Bianchi
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.
| |
Collapse
|
7
|
Shu Y, Xiao B, Wu Q, Liu T, Du Y, Tang H, Chen S, Feng L, Long L, Li Y. The Ephrin-A5/EphA4 Interaction Modulates Neurogenesis and Angiogenesis by the p-Akt and p-ERK Pathways in a Mouse Model of TLE. Mol Neurobiol 2014; 53:561-576. [PMID: 25502292 DOI: 10.1007/s12035-014-9020-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/18/2014] [Indexed: 02/07/2023]
Abstract
Studies have shown that neurogenesis and angiogenesis do exist in temporal lobe epilepsy (TLE). The ephrin ligands and Eph receptors are the largest members of receptor tyrosine kinases, and their interaction via cell-cell contact participates in cell proliferation, differentiation, migration, and tissue remodeling. However, there is little information about the function of the ephrin-A5/EphA4 complex in TLE. In the current study, we found that ephrin-A5 was expressed in astrocytes, while EphA4 existed in endothelial cells in the hippocampus in a mouse model of TLE. Furthermore, the messenger RNA (mRNA) and protein levels of both ephrin-A5 and EphA4 and the binding capacity of ephrin-A5/EphA4 showed gradual increase in spatiotemporal course. When ephrin-A5-Fc was injected into the hippocampus at 3 days post-status epilepticus (SE) for 7 days, the spontaneous recurrent seizure (SRS) frequency and intensity of the mice attenuated in the following 2 weeks. Furthermore, doublecortin-positive neuronal progenitor cells were reduced in the subgranular zone, and the density of microvessels decreased in the hilus. The molecular mechanism was attributed to ephrin-A5-Fc-induced inhibition of phosphorylated ERK (p-ERK) and phosphorylated Akt (p-Akt), and also EphA4 and VEGF reduction. In summary, interaction between ephrin-A5 and EphA4 could mediate the ERK and Akt signaling pathways in pilocarpine-induced epilepsy, and intervention of the ephrin/Eph interaction may play an essential role in the suppression of newborn neuron generation, microvessel remodeling, and SRS in a mouse model of TLE. The ephrin-A5/EphA4 communication may provide a potential therapy for the treatment of TLE.
Collapse
Affiliation(s)
- Yi Shu
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China.
| | - Qian Wu
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Tiantian Liu
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Yang Du
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Haiyun Tang
- Department of Radiology, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Si Chen
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Yi Li
- Department of Neurology, Xiangya Hospital, Central South University, Hunan, 410008, China.
- Department of Neurology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA, 01604, USA.
| |
Collapse
|
8
|
Lu M, Lawrence DA, Marsters S, Acosta-Alvear D, Kimmig P, Mendez AS, Paton AW, Paton JC, Walter P, Ashkenazi A. Opposing unfolded-protein-response signals converge on death receptor 5 to control apoptosis. Science 2014; 345:98-101. [PMID: 24994655 PMCID: PMC4284148 DOI: 10.1126/science.1254312] [Citation(s) in RCA: 455] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Protein folding by the endoplasmic reticulum (ER) is physiologically critical; its disruption causes ER stress and augments disease. ER stress activates the unfolded protein response (UPR) to restore homeostasis. If stress persists, the UPR induces apoptotic cell death, but the mechanisms remain elusive. Here, we report that unmitigated ER stress promoted apoptosis through cell-autonomous, UPR-controlled activation of death receptor 5 (DR5). ER stressors induced DR5 transcription via the UPR mediator CHOP; however, the UPR sensor IRE1α transiently catalyzed DR5 mRNA decay, which allowed time for adaptation. Persistent ER stress built up intracellular DR5 protein, driving ligand-independent DR5 activation and apoptosis engagement via caspase-8. Thus, DR5 integrates opposing UPR signals to couple ER stress and apoptotic cell fate.
Collapse
Affiliation(s)
- Min Lu
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - David A Lawrence
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Scot Marsters
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Diego Acosta-Alvear
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA.Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Philipp Kimmig
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA.Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Aaron S Mendez
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA.Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, South Australia, 5005, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, South Australia, 5005, Australia
| | - Peter Walter
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA.Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA.
| | - Avi Ashkenazi
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
9
|
Lu M, Marsters S, Ye X, Luis E, Gonzalez L, Ashkenazi A. E-cadherin couples death receptors to the cytoskeleton to regulate apoptosis. Mol Cell 2014; 54:987-998. [PMID: 24882208 DOI: 10.1016/j.molcel.2014.04.029] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 02/11/2014] [Accepted: 04/22/2014] [Indexed: 01/14/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a cellular process essential to the development and maintenance of solid tissues. In cancer, EMT suppresses apoptosis, but the mechanisms remain unclear. EMT selectively attenuated apoptosis signaling via the death receptors DR4 and DR5. Loss of the epithelial cell adhesion protein E-cadherin recapitulated this outcome, whereas homotypic E-cadherin engagement promoted apoptotic signaling via DR4/DR5, but not Fas. Depletion of α-catenin, which couples E-cadherin to the actin cytoskeleton, or actin polymerization inhibitors similarly attenuated DR4/DR5-induced apoptosis. E-cadherin bound specifically to ligated DR4/DR5, requiring extracellular cadherin domain 1 and calcium. E-cadherin augmented DR4/DR5 clustering and assembly of the death-inducing signaling complex (DISC), increasing caspase-8 activation in high molecular weight cell fractions. Conversely, EMT attenuated DR4/DR5-mediated DISC formation and caspase-8 stimulation. Consistent with these findings, epithelial cancer cell lines expressing higher E-cadherin levels displayed greater sensitivity to DR4/DR5-mediated apoptosis. These results have potential implications for tissue homeostasis as well as cancer therapy.
Collapse
Affiliation(s)
- Min Lu
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Scot Marsters
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Xiaofen Ye
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Elizabeth Luis
- Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lino Gonzalez
- Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Avi Ashkenazi
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
10
|
Shimoni M, Herschhorn A, Britan-Rosich Y, Kotler M, Benhar I, Hizi A. The isolation of novel phage display-derived human recombinant antibodies against CCR5, the major co-receptor of HIV. Viral Immunol 2014; 26:277-90. [PMID: 23941674 DOI: 10.1089/vim.2012.0029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Selecting for antibodies against specific cell-surface proteins is a difficult task due to many unrelated proteins that are expressed on the cell surface. Here, we describe a method to screen antibody-presenting phage libraries against native cell-surface proteins. We applied this method to isolate antibodies that selectively recognize CCR5, which is the major co-receptor for HIV entry (consequently, playing a pivotal role in HIV transmission and pathogenesis). We employed a phage screening strategy by using cells that co-express GFP and CCR5, along with an excess of control cells that do not express these proteins (and are otherwise identical to the CCR5-expressing cells). These control cells are intended to remove most of the phages that bind the cells nonspecifically; thus leading to an enrichment of the phages presenting anti-CCR5-specific antibodies. Subsequently, the CCR5-presenting cells were quantitatively sorted by flow cytometry, and the bound phages were eluted, amplified, and used for further successive selection rounds. Several different clones of human single-chain Fv antibodies that interact with CCR5-expressing cells were identified. The most specific monoclonal antibody was converted to a full-length IgG and bound the second extracellular loop of CCR5. The experimental approach presented herein for screening for CCR5-specific antibodies can be applicable to screen antibody-presenting phage libraries against any cell-surface expressed protein of interest.
Collapse
Affiliation(s)
- Moria Shimoni
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
11
|
Lyon CA, Johnson JL, White S, Sala-Newby GB, George SJ. EC4, a truncation of soluble N-cadherin, reduces vascular smooth muscle cell apoptosis and markers of atherosclerotic plaque instability. Mol Ther Methods Clin Dev 2014; 1:14004. [PMID: 26015951 PMCID: PMC4362368 DOI: 10.1038/mtm.2014.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/05/2014] [Indexed: 11/21/2022]
Abstract
Atherosclerotic plaque instability is precipitated by vascular smooth muscle cell apoptosis in the fibrous cap, weakening it and leading to plaque rupture. We previously showed that reducing smooth muscle cell apoptosis with soluble N-cadherin (SNC) increased features of plaque stability. We have now identified the active site of SNC and examined whether a truncated form containing this site retains the antiapoptotic effect. SNC was mutated to prevent interaction with N-cadherin or fibroblast growth factor receptor (FGFR). Interaction with FGFR in the extracellular (EC) 4 domain of SNC was essential for the antiapoptotic effect. Therefore, we made a truncated form consisting of the EC4 domain. EC4 significantly reduced smooth muscle cell, macrophage, and endothelial cell apoptosis in vitro by ~70%, similar to SNC. Elevation of plasma levels of EC4 in male apolipoprotein E-deficient mice with existing atherosclerosis significantly reduced apoptosis in brachiocephalic artery plaques by ~50%. EC4 reduced plaque size and the incidence of buried fibrous layers and the macrophage:smooth muscle cell ratio (surrogate markers of plaque instability). Interaction of EC4 with FGFR induced potent antiapoptotic signaling in vitro and in vivo. EC4 modulates atherosclerosis in mice demonstrating its therapeutic potential for retarding plaque size and instability.
Collapse
Affiliation(s)
- Cressida A Lyon
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Jason L Johnson
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Stephen White
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | | | - Sarah J George
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| |
Collapse
|
12
|
Double-chimera proteins to enhance recruitment of endothelial cells and their progenitor cells. Int J Cardiol 2013; 167:1560-9. [DOI: 10.1016/j.ijcard.2012.04.094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 03/27/2012] [Accepted: 04/14/2012] [Indexed: 11/15/2022]
|
13
|
A long-acting erythropoietin fused with noncytolytic human Fc for the treatment of anemia. Arch Pharm Res 2013; 35:757-9. [PMID: 22644842 DOI: 10.1007/s12272-012-0500-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The Fc fusion technology has been introduced to generate long-acting antagonistic drugs such as Enbrel, Orencia and Amevive. Here, Genexine created a novel noncytolytic hybrid Fc (hyFc) as a carrier of agonistic protein drugs using naturally existing IgD and IgG4 Fcs without any mutation in the hyFc region. The erythropoietin (EPO) fused with hyFc exhibited little binding activity to FcγR and C1q molecules that are main mediators for death of target cells. The EPO-hyFc showed higher in vitro and in vivo bioactivities than EPOIgG1 Fc and highly glycosylated EPO (Aranesp). Phase I clinical trial with EPO-hyFc is currently undergoing in Korea.
Collapse
|
14
|
Abstract
TNF-α-blocking agents such as infliximab, adalimumab and etanercept are widely used for the treatment of severe inflammatory diseases including rheumatoid arthritis and psoriasis. The currently used TNF-α blockers have Fc regions of the human IgG1 subtype, which is advantageous in terms of in vivo half-life but also raise the potential for unwanted effector-mediated effects, such as antibody dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC). To address this issue, we constructed a novel hybrid protein by fusing the TNF receptor (TNFR) with a hybrid Fc (hyFc) consisting of the CH2 and CH3 regions of IgG4 and the highly flexible hinge regions of IgD which would not have ADCC and CDC activity. The resulting fusion protein, TNFR-hyFc, was over-expressed in CHO and pharmacological characteristics were evaluated in comparison with the structurally similar etanercept. TNFR-hyFc effectively neutralized TNF-α in L929 bioassay and showed a 1.5-fold higher neutralizing activity compared to etanercept. In a pharmacokinetic study in cynomolgus monkeys, TNFR-hyFc showed plasma half-life and AUC comparable to etanercept. In a mouse collagen induced arthritis model, TNFR-hyFc showed significant amelioration of arthritis compared to etanercept or vehicle control. In an LPS-induced septic shock model, TNFR-hyFc showed a similar level of protection against mortality as etanercept. These results confirm the feasibility of the TNFR-hyFc as an effective TNF-α blocker for the treatment of inflammatory diseases.
Collapse
|
15
|
Lee JH, Yeo J, Park HS, Sung G, Lee SH, Yang SH, Sung YC, Kang JH, Park CS. Biochemical characterization of a new recombinant TNF receptor-hyFc fusion protein expressed in CHO cells. Protein Expr Purif 2012; 87:17-26. [PMID: 23009882 DOI: 10.1016/j.pep.2012.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 08/27/2012] [Accepted: 09/07/2012] [Indexed: 12/31/2022]
Abstract
The currently used Tumor Nectosis Factor (TNF)-α blockers such as infliximab, adalimumab and etanercept have Fc regions of the human IgG1 subtype have advantages in terms of in vivo half-life, however these could raise potential concerns for unwanted effector-mediated effects, such as antibody dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC). To address this issue, we constructed a novel hybrid protein with decreased ADCC and CDC potentials by fusing the TNF receptor to a hybrid Fc (hyFc) containing CH2 and CH3 regions of IgG4 and highly flexible hinge regions of IgD which neither has ADCC and CDC activities. The resulting fusion protein, TNFR-hyFc, was over-expressed in CHO cells. For use as a pre-clinical material in pharmacology, PK and toxicological evaluations were carried out for biochemical characterization which was then compared with etanercept that has similarity in structure. Amino acid composition analysis and peptide mapping showed that the expressed TNFR-hyFc matched the theoretical composition derived from the DNA sequence. Matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) showed that TNFR-hyFc is 2.9 kDa larger than etanercept. MALDI-TOF after removal of N-glycans by PNGase treatment showed that TNFR-hyFc is 3.9 kDa larger than etanercept. Isoelectric focusing and monosaccharide analysis showed that TNFR-hyFc is slightly more acidic than etanercept. N-terminal amino acid sequencing showed that N-terminal heterogeneity is present in both TNFR-hyFc and etanercept, although the ratios are somewhat different. Glycan analysis showed that the main glycan form is bi-antennary, similar to etanercept.
Collapse
Affiliation(s)
- Jung-Hwan Lee
- Biopharmaceutical Research Labs, Research Center, Dong-A Pharmaceutical Co., Ltd., Sanggal-dong, Giheung-gu, Yongin-si, Kyungki-do 446-905, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Im SJ, Yang SI, Yang SH, Choi DH, Choi SY, Kim HS, Jang DS, Jin KS, Chung YK, Kim SH, Paik SH, Park YC, Chung MK, Kim YB, Han KH, Choi KY, Sung YC. Natural form of noncytolytic flexible human Fc as a long-acting carrier of agonistic ligand, erythropoietin. PLoS One 2011; 6:e24574. [PMID: 21957455 PMCID: PMC3174958 DOI: 10.1371/journal.pone.0024574] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 08/14/2011] [Indexed: 12/27/2022] Open
Abstract
Human IgG1 Fc has been widely used as a bioconjugate, but exhibits shortcomings, such as antibody- and complement-mediated cytotoxicity as well as decreased bioactivity, when applied to agonistic proteins. Here, we constructed a nonimmunogenic, noncytolytic and flexible hybrid Fc (hyFc) consisting of IgD and IgG4, and tested its function using erythropoietin (EPO) conjugate, EPO-hyFc. Despite low amino acid homology (20.5%) between IgD Fc and IgG4 Fc, EPO-hyFc retained “Y-shaped” structure and repeated intravenous administrations of EPO-hyFc into monkeys did not generate EPO-hyFc-specific antibody responses. Furthermore, EPO-hyFc could not bind to FcγR I and C1q in contrast to EPO-IgG1 Fc. In addition, EPO-hyFc exhibited better in vitro bioactivity and in vivo bioactivity in rats than EPO-IgG1 Fc, presumably due to the high flexibility of IgD. Moreover, the mean serum half-life of EPO-hyFc(H), a high sialic acid content form of EPO-hyFc, was approximately 2-fold longer than that of the heavily glycosylated EPO, darbepoetin alfa, in rats. More importantly, subcutaneous injection of EPO-hyFc(H) not only induced a significantly greater elevation of serum hemoglobin levels than darbepoetin alfa in both normal rats and cisplatin-induced anemic rats, but also displayed a delayed time to maximal serum level and twice final area-under-the-curve (AUClast). Taken together, hyFc might be a more attractive Fc conjugate for agonistic proteins/peptides than IgG1 Fc due to its capability to elongate their half-lives without inducing host effector functions and hindering bioactivity of fused molecules. Additionally, a head-to-head comparison demonstrated that hyFc-fusion strategy more effectively improved the in vivo bioactivity of EPO than the hyperglycosylation approach.
Collapse
Affiliation(s)
- Se Jin Im
- Division of Molecular and Life Sciences, POSTECH, Pohang, Republic of Korea
| | - Sang In Yang
- Research Institute, Genexine Co., Seongnam, Republic of Korea
| | - Se Hwan Yang
- Research Institute, Genexine Co., Seongnam, Republic of Korea
| | - Dong Hoon Choi
- Division of Molecular and Life Sciences, POSTECH, Pohang, Republic of Korea
| | - So Young Choi
- Research Institute, Genexine Co., Seongnam, Republic of Korea
| | - Hea Sook Kim
- Research Institute, Genexine Co., Seongnam, Republic of Korea
| | - Do Soo Jang
- Research Institute, Genexine Co., Seongnam, Republic of Korea
| | - Kyeong Sik Jin
- Pohang Accelerator Laboratory, POSTECH, Pohang, Republic of Korea
| | - Yo-Kyung Chung
- Central Research Institute, Green Cross Co., Yongin, Republic of Korea
| | - Seung-Hee Kim
- Central Research Institute, Green Cross Co., Yongin, Republic of Korea
| | - Sang Hoon Paik
- Central Research Institute, Green Cross Co., Yongin, Republic of Korea
| | - Yoo Chang Park
- Central Research Institute, Green Cross Co., Yongin, Republic of Korea
| | - Moon Koo Chung
- Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Daejon, Republic of Korea
| | - Yong Bum Kim
- Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Daejon, Republic of Korea
| | - Kang-Hyun Han
- Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Daejon, Republic of Korea
| | - Kwan Yong Choi
- Division of Molecular and Life Sciences, POSTECH, Pohang, Republic of Korea
| | - Young Chul Sung
- Division of Molecular and Life Sciences, POSTECH, Pohang, Republic of Korea
- Research Institute, Genexine Co., Seongnam, Republic of Korea
- * E-mail:
| |
Collapse
|
17
|
Konduru K, Bradfute SB, Jacques J, Manangeeswaran M, Nakamura S, Morshed S, Wood SC, Bavari S, Kaplan GG. Ebola virus glycoprotein Fc fusion protein confers protection against lethal challenge in vaccinated mice. Vaccine 2011; 29:2968-77. [PMID: 21329775 PMCID: PMC3070761 DOI: 10.1016/j.vaccine.2011.01.113] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 01/26/2011] [Accepted: 01/31/2011] [Indexed: 01/09/2023]
Abstract
Ebola virus is a Filoviridae that causes hemorrhagic fever in humans and induces high morbidity and mortality rates. Filoviruses are classified as "Category A bioterrorism agents", and currently there are no licensed therapeutics or vaccines to treat and prevent infection. The Filovirus glycoprotein (GP) is sufficient to protect individuals against infection, and several vaccines based on GP are under development including recombinant adenovirus, parainfluenza virus, Venezuelan equine encephalitis virus, vesicular stomatitis virus (VSV) and virus-like particles. Here we describe the development of a GP Fc fusion protein as a vaccine candidate. We expressed the extracellular domain of the Zaire Ebola virus (ZEBOV) GP fused to the Fc fragment of human IgG1 (ZEBOVGP-Fc) in mammalian cells and showed that GP undergoes the complex furin cleavage and processing observed in the native membrane-bound GP. Mice immunized with ZEBOVGP-Fc developed T-cell immunity against ZEBOV GP and neutralizing antibodies against replication-competent VSV-G deleted recombinant VSV containing ZEBOV GP. The ZEBOVGP-Fc vaccinated mice were protected against challenge with a lethal dose of ZEBOV. These results show that vaccination with the ZEBOVGP-Fc fusion protein alone without the need of a viral vector or assembly into virus-like particles is sufficient to induce protective immunity against ZEBOV in mice. Our data suggested that Filovirus GP Fc fusion proteins could be developed as a simple, safe, efficacious, and cost effective vaccine against Filovirus infection for human use.
Collapse
Affiliation(s)
- Krishnamurthy Konduru
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Nagashima H, Kaneko K, Yamanoi A, Motoi S, Konakahara S, Kohroki J, Masuho Y. TNF receptor II fusion protein with tandemly repeated Fc domains. J Biochem 2011; 149:337-46. [PMID: 21278157 DOI: 10.1093/jb/mvq149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The extracellular domain of tumour necrosis factor (TNF) receptor II fused with the human IgG1 Fc region (TNFRII-Fc), as well as antibodies against TNF, has been used to treat rheumatoid arthritis. However, TNFRII-Fc is less effective than these antibodies in terms of antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against cells bearing TNF on the cell surface. We hypothesized that these activities could be increased by fusing TNFRII with tandemly repeated Fc (TNFRII-Fc-Fc). The affinities of TNFRII-Fc-Fc for soluble TNF-α and transmembrane TNF-α and the TNF-α cytotoxicity-inhibitory activity were as potent as those of TNFRII-Fc. TNFRII-Fc-Fc showed much higher binding avidity for Fcγ receptors than TNFRII-Fc and was more potent in terms of both ADCC and CDC against cells expressing transmembrane TNF-α. TNFRII-Fc-Fc of 80 kDa, as well as TNFRII-Fc-Fc of 200 kDa, was detected. TNFRII-Fc-Fc (80 kDa) was as potent as TNFRII-Fc in terms of both ADCC and CDC. These results suggest that Fc multimerization of receptor-Fc fusion proteins can augment effector functions such as ADCC and CDC, and thereby have the potential to provide a superior therapeutic effect. This may be the case not only for TNFRII-Fc but also for other receptor-Fc fusion proteins.
Collapse
Affiliation(s)
- Hiroaki Nagashima
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Yamazaki 2641, Noda, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
19
|
Wright GJ. Signal initiation in biological systems: the properties and detection of transient extracellular protein interactions. MOLECULAR BIOSYSTEMS 2010; 5:1405-12. [PMID: 19593473 PMCID: PMC2898632 DOI: 10.1039/b903580j] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Extracellular glycoprotein interactions are not detected by most high throughput assays creating “blind-spots” in protein interaction maps. This review examines this problem and discusses recent advances that have begun to address it.
Individual cells within biological systems frequently coordinate their functions through signals initiated by specific extracellular protein interactions involving receptors that bridge the cellular membrane. Due to their biochemical nature, these membrane-embedded receptor proteins are difficult to manipulate and their interactions are characterised by very weak binding strengths that cannot be detected using popular high throughput assays. This review will provide a general outline of the biochemical attributes of receptor proteins focussing in particular on the biophysical properties of their transient interactions. Methods that are able to detect these weak extracellular binding events and especially those that can be used for identifying novel interactions will be compared. Finally, I discuss the feasibility of constructing a complete and accurate extracellular protein interaction map, and the methods that are likely to be useful in achieving this goal.
Collapse
Affiliation(s)
- Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.
| |
Collapse
|
20
|
Myllymaa S, Pasternack A, Mottershead DG, Poutanen M, Pulkki MM, Pelliniemi LJ, Ritvos O, Laitinen MPE. Inhibition of oocyte growth factors in vivo modulates ovarian folliculogenesis in neonatal and immature mice. Reproduction 2009; 139:587-98. [PMID: 20007639 DOI: 10.1530/rep-09-0391] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Growth differentiation factor-9 (GDF9) and bone morphogenetic protein-15 (BMP15) are among the key regulators transmitting the signaling between the oocyte and the surrounding granulosa cells. Previously, it has been shown that a recombinant BMP type II receptor ectodomain-Fc fusion protein (BMPR2ecd-Fc) is able to inhibit the actions of GDF9 and BMP15 in vitro. Here, we have produced bioactive BMPR2ecd-Fc, which was injected i.p. into neonatal mice. Early folliculogenesis was first studied by injecting mice five times with various doses of BMPR2ecd-Fc during the postnatal days 4-12. Folliculogenesis was affected dose dependently, as evidenced by a decreased mitogenesis of granulosa cells of the growing follicles. Furthermore, we also noticed a decrease in the number of secondary and tertiary follicles as well as an increase in the oocyte size. Electron microscopic analysis revealed that the ultrastructure of the granulosa cells of the primary follicles was not affected by the BMPR2ecd-Fc treatment. A second study was conducted to investigate whether a longer treatment with 12 injections during postnatal days 4-28 would inhibit folliculogenesis. Similar effects were observed in the two studies on the early follicular developmental stages. However, in the long-term study, later stages of folliculogenesis were not blocked but rather increased numbers of antral follicles, preovulatory follicles, and corpora lutea were found. We conclude that BMPR2ecd-Fc is a potent modulator of ovarian folliculogenesis in vivo, and thus, is a valuable tool for studying the physiology and downstream effects of oocyte-derived growth factors in vivo.
Collapse
Affiliation(s)
- Samu Myllymaa
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Huang YS, Wen XF, Wu YL, Wang YF, Fan M, Yang ZY, Liu W, Zhou LF. Engineering a pharmacologically superior form of granulocyte-colony-stimulating factor by fusion with gelatin-like-protein polymer. Eur J Pharm Biopharm 2009; 74:435-41. [PMID: 19995603 DOI: 10.1016/j.ejpb.2009.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 12/01/2009] [Accepted: 12/02/2009] [Indexed: 10/20/2022]
Abstract
The plasma half-life of therapeutic proteins is a critical factor in many clinical applications. Therefore, new strategies to prolong plasma half-life of long-acting peptides and protein drugs are in high demand. Here, we designed an artificial gelatin-like protein (GLK) and fused this hydrophilic GLK polymer to granulocyte-colony-stimulating factor (G-CSF) to generate a chimeric GLK/G-CSF fusion protein. The genetically engineered recombinant GLK/G-CSF (rGLK/G-CSF) fusion protein was purified from Pichia pastoris. In vitro studies demonstrated that rGLK/G-CSF possessed an enlarged hydrodynamic radius, improved thermal stability and retained full bioactivity compared to unfused G-CSF. Following a single subcutaneous administration to rats, the rGLK/G-CSF fusion protein displayed a slower plasma clearance rate and stimulated greater and longer lasting increases in circulating white blood cells than G-CSF. Our findings indicate that fusion with this artificial, hydrophilic, GLK polymer provides many advantages in the construction of a potent hematopoietic factor with extended plasma half-life. This approach could be easily applied to other therapeutic proteins and have important clinical applications.
Collapse
Affiliation(s)
- Yan-Shan Huang
- Department of Cell Biology, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Otani T, Hashizume T, Nagaoka T, Fukuda T, Tang CK, Salomon DS, Seno M. Production of biologically active IgG hinge-tag soluble epidermal growth factor receptors (ErbB). Biotechnol Lett 2009; 32:361-6. [PMID: 19898750 DOI: 10.1007/s10529-009-0160-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 10/18/2009] [Accepted: 10/22/2009] [Indexed: 10/20/2022]
Abstract
The extracellular domains (ECD) of epidermal growth factor receptors, ErbB1, 2, 3 and 4, were designed as soluble dimeric forms. Each ECD was fused to a short hinge region derived from IgG, such that the stable dimer could be formed with disulfide bridges. This hinge-tagged design minimized the molecular weight to approximately 50% of the conventional Fc-fusion design without an Fc domain of IgG. The refolded dimers could be easily analyzed and characterized by SDS-PAGE. Hinge-tagged soluble ErbBs demonstrated significant affinity for betacellulin and heregulin. The IgG hinge-tag should be a simple method to design soluble dimers that would be useful for high throughput screening of ligands, antagonists or derivatives.
Collapse
Affiliation(s)
- Takayuki Otani
- Department of Medical and Bioengineering Science, Graduate School of Natural Science and Technology, Okayama University, 3.1.1 Tsushima-Naka, Kita-ku, Okayama, 700-8530, Japan
| | | | | | | | | | | | | |
Collapse
|
23
|
Terai M, Tamura Y, Alexeev V, Ohtsuka E, Berd D, Mastrangelo MJ, Sato T. Human interleukin 10 receptor 1/IgG1-Fc fusion proteins: immunoadhesins for human IL-10 with therapeutic potential. Cancer Immunol Immunother 2009; 58:1307-17. [PMID: 19142637 PMCID: PMC11030067 DOI: 10.1007/s00262-008-0644-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Accepted: 12/08/2008] [Indexed: 10/21/2022]
Abstract
Interleukin 10 (IL-10) is produced by various types of human cancer, including malignant melanoma, and plays an important role in negative regulation of cell-mediated immune responses against tumors. We have developed chimeric molecules (immunoadhesins), combining the extracellular domain of human interleukin 10 receptor 1 (IL-10R1) with the Fc regions of human IgG1 heavy chain and investigated their capability of blocking the biological activities of human IL-10. Monomeric and dimeric immunoadhesins (IL-10R1/IgG1) constructs were tested for capturing human IL-10 and blocking its biological activities. Plasmid vectors that contained the IL-10 immunoadhesin constructs were directly transfected into human melanoma cell lines. Transfection of plasmid vectors into melanoma cell lines resulted in capturing of exogenously added as well as endogeneously produced IL-10. The supernatants obtained from an IL-10 non-producing melanoma cell line transfected with monomeric IL-10 immunoadhesin plasmids most efficiently captured exogenously added IL-10, compared to those obtained with the dimeric IL-10R1/IgG1 plasmid vector. Transfection of IL-10-producing melanoma cells with the monomeric IL-10 immunoadhesin plasmids totally captured endogenously produced IL-10 and enhanced T cell responses against allogeneic melanoma cells. Furthermore, purified monomeric IL-10 immunoadhesin protein showed IL-10 capturing efficacy compatible with that of IL-10-specific monoclonal antibodies. Collectively, these studies indicate that IL-10 immunoadhesins, especially in monomeric form, are potent inhibitors of biological activities of IL-10 and suggest that these molecules, alone or in conjunctions with other immunotherapeutic approaches, can be utilized for the immuno-targeting of IL-10 producing tumors.
Collapse
Affiliation(s)
- Mizue Terai
- Jefferson Medical College of Thomas Jefferson University, 1015 Walnut Street, Suite 1024, Philadelphia, PA 19107-5099 USA
- Chiba University, Chiba, Japan
| | | | - Vitali Alexeev
- Jefferson Medical College of Thomas Jefferson University, 1015 Walnut Street, Suite 1024, Philadelphia, PA 19107-5099 USA
| | | | - David Berd
- Jefferson Medical College of Thomas Jefferson University, 1015 Walnut Street, Suite 1024, Philadelphia, PA 19107-5099 USA
| | - Michael J. Mastrangelo
- Jefferson Medical College of Thomas Jefferson University, 1015 Walnut Street, Suite 1024, Philadelphia, PA 19107-5099 USA
| | - Takami Sato
- Jefferson Medical College of Thomas Jefferson University, 1015 Walnut Street, Suite 1024, Philadelphia, PA 19107-5099 USA
| |
Collapse
|
24
|
Paoletti F, Covaceuszach S, Konarev PV, Gonfloni S, Malerba F, Schwarz E, Svergun DI, Cattaneo A, Lamba D. Intrinsic structural disorder of mouse proNGF. Proteins 2009; 75:990-1009. [PMID: 19089979 DOI: 10.1002/prot.22311] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The unprocessed precursor of the Nerve Growth Factor (NGF), proNGF, has additional functions, besides its initially described role as a chaperone for NGF folding. The precursor protein endows apoptotic and/or neurotrophic properties, in contrast to the mature part. The structural and molecular basis for such distinct activities are presently unknown. Aiming to gain insights into the specific molecular interactions that govern rm-proNGF biological activities versus those of its mature counterpart, a structural study by synchrotron small angle X-ray scattering (SAXS) in solution was carried out. The different binding properties of the two proteins were investigated by surface plasmon resonance (SPR) using, as structural probes, a panel of anti-NGF antibodies and the soluble forms of TrkA and p75(NTR) receptors. SAXS measurements revealed the rm-proNGF to be dimeric and anisometric, with the propeptide domain being intrinsically unstructured. Ab initio reconstructions assuming twofold symmetry generated two types of structural models, a globular "crab-like" and an elongated shape that resulted in equally good fits of the scattering data. A novel method accounting for possible coexistence of different conformations contributing to the experimental scattering pattern, with no symmetry constraints, suggests the "crab-like" to be a more likely proNGF conformation. To exploit the potential of chemical stabilizers affecting the existing conformational protein populations, SAXS data were also collected in the presence of ammonium sulphate. An increase of the proNGF compactness was observed. SPR data pinpoints that the propeptide of proNGF may act as an intrinsically unstructured protein domain, characterized by a molecular promiscuity in the interaction/binding to multiple partners (TrkA and p75(NTR) receptors and a panel of neutralizing anti-NGF antibodies) depending on the physiological conditions of the cell. These data provide a first insight into the structural basis for the selectivity of mouse short proNGF, versus NGF, towards its binding partners.
Collapse
Affiliation(s)
- Francesca Paoletti
- SISSA-ISAS, Building Q1, Area Science Park - Basovizza, S.S 14 Km 163.5, 34012 Trieste, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Tijink BM, Laeremans T, Budde M, Stigter-van Walsum M, Dreier T, de Haard HJ, Leemans CR, van Dongen GAMS. Improved tumor targeting of anti-epidermal growth factor receptor Nanobodies through albumin binding: taking advantage of modular Nanobody technology. Mol Cancer Ther 2008; 7:2288-97. [PMID: 18723476 DOI: 10.1158/1535-7163.mct-07-2384] [Citation(s) in RCA: 228] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The approximately 15-kDa variable domains of camelid heavy-chain-only antibodies (called Nanobodies) can easily be formatted as multivalent or multispecific single-chain proteins. Because of fast excretion, however, they are less suitable for therapy of cancer. In this study, we aimed for improved tumor targeting of a bivalent anti-epidermal growth factor receptor (EGFR) Nanobody (alphaEGFR-alphaEGFR) by fusion to a Nanobody unit binding to albumin (alphaAlb). Biodistributions of alphaEGFR-alphaEGFR, alphaEGFR-alphaEGFR-alphaAlb ( approximately 50 kDa), alphaTNF-alphaTNF-alphaAlb (control, binding tumor necrosis factor-alpha), and the approximately 150-kDa anti-EGFR antibody cetuximab were compared in A431 xenograft-bearing mice. The proteins were radiolabeled with (177)Lu to facilitate quantification. Tumor uptake of (177)Lu-alphaEGFR-alphaEGFR decreased from 5.0 +/- 1.4 to 1.1 +/- 0.1 %ID/g between 6 and 72 h after injection. Due to its rapid blood clearance, tumor-to-blood ratios >80 were obtained within 6 h after injection. Blood clearance became dramatically slower and tumor uptake became significantly higher by introduction of alphaAlb. Blood levels of alphaEGFR-alphaEGFR-alphaAlb were 21.2 +/- 2.5, 11.9 +/- 0.6, and 4.0 +/- 1.4 and tumor levels were 19.4 +/- 5.5, 35.2 +/- 7.5, and 28.0 +/- 6.8 %ID/g at 6, 24, and 72 h after injection, respectively. Tumor uptake was at least as high as for cetuximab (15.5 +/- 3.9, 27.1 +/- 7.9, and 25.6 +/- 6.1 %ID/g) and significantly higher than for alphaTNF-alphaTNF-alphaAlb. alphaEGFR-alphaEGFR-alphaAlb showed faster and deeper tumor penetration than cetuximab. These data show that simple fusion of alphaEGFR and alphaAlb building blocks results in a bifunctional Nanobody format, which seems more favorable for therapy as far as pharmacokinetics and tumor deposition are concerned.
Collapse
Affiliation(s)
- Bernard M Tijink
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Cowan FM. Staphylococcus Aureus Protein A Immunoadsorption: The Rationale for Systemic Protein A Immunotherapy. ACTA ACUST UNITED AC 2008. [DOI: 10.1080/105172397243277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
27
|
Nagaoka T, Fukuda T, Hashizume T, Nishiyama T, Tada H, Yamada H, Salomon DS, Yamada S, Kojima I, Seno M. A betacellulin mutant promotes differentiation of pancreatic acinar AR42J cells into insulin-producing cells with low affinity of binding to ErbB1. J Mol Biol 2008; 380:83-94. [PMID: 18508082 DOI: 10.1016/j.jmb.2008.03.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 03/10/2008] [Accepted: 03/25/2008] [Indexed: 01/08/2023]
Abstract
Betacellulin (BTC) is one of the members of the epidermal growth factor (EGF) ligand family of ErbB receptor tyrosine kinases. It is a differentiation factor as well as a potent mitogen. BTC promotes the differentiation of pancreatic acinar-derived AR42J cells into insulin-producing cells. It independently and preferentially binds to two type I tyrosine kinase receptors, the EGF receptor (ErbB1) and ErbB4. However, the physiochemical characteristics of BTC that are responsible for its preferential binding to these two receptors have not been fully defined. In this study, to investigate the essential amino acid residues of BTC for binding to the two receptors, we introduced point mutations into the EGF domain of BTC employing error-prone PCR. The receptor binding abilities of 190 mutants expressed in Escherichia coli were assessed by enzyme immunoassay. Replacement of the glutamic acid residue at position 88 with a lysine residue in BTC was found to produce a significant loss of affinity for binding to ErbB1, while the affinity of binding to ErbB4 was unchanged. In addition, the mutant of BTC-E/88/K showed less growth-promoting activity on BALB/c 3T3 cells compared with that of the wild-type BTC protein. Interestingly, the BTC mutant protein promoted differentiation of pancreatic acinar AR42J cells at a high frequency into insulin-producing cells compared with AR42J cells that were treated with wild-type BTC protein. These results indicate the possibility of designing BTC mutants, which have an activity of inducing differentiation only, without facilitating growth promotion.
Collapse
Affiliation(s)
- Tadahiro Nagaoka
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-Naka, Okayama 700-8530, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sung BH, Kim SH, Yeo MG, Kim JK, Song WK. Human soluble E-selectin immunoadhesin inhibits leukemic monocyte adhesion to endothelial cells. Cell Biochem Funct 2007; 25:585-9. [PMID: 16892455 DOI: 10.1002/cbf.1361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Immunoadhesins are immunoglobulin (Ig)-like chimeric proteins comprised of target-binding regions fused to the Fc-hinge region of Ig, and are designed to have a long half-life and antibody-like properties. In an effort to find a good candidate for therapeutic use for inflammatory responses, we constructed a soluble human E-selectin immunoadhesin containing the extracellular region of human E-selectin fused to the Fc-hinge region of human IgG, and determined its effects on leukocyte adhesion and rolling in vitro. Our results revealed that the adhesion of leukocytes to endothelial cells was efficiently inhibited in the presence of 50 nM E-selectin immunoadhesin. In addition, the E-selectin immunoadhesin significantly inhibited leukocyte rolling on endothelial cells in perfusion experiments performed at 1.0 dyne/cm(2) wall shear stress. These findings indicate that our E-selectin immunoadhesin decreases leukocyte attachment and rolling in vitro, suggesting that this immunoadhesin may be a promising candidate for therapeutic anti-inflammatory use.
Collapse
Affiliation(s)
- Bong Hwan Sung
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | | | | | | | | |
Collapse
|
29
|
Huston A, Lyman GH. Agents under investigation for the treatment and prevention of neutropenia. Expert Opin Investig Drugs 2007; 16:1831-40. [DOI: 10.1517/13543784.16.11.1831] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
30
|
McKenna SD, Feger G, Kelton C, Yang M, Ardissone V, Cirillo R, Vitte PA, Jiang X, Campbell RK. Tumor necrosis factor (TNF)-soluble high-affinity receptor complex as a TNF antagonist. J Pharmacol Exp Ther 2007; 322:822-8. [PMID: 17495128 DOI: 10.1124/jpet.107.119875] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A novel high-affinity inhibitor of tumor necrosis factor (TNF) is described, which is created by the fusion of the extracellular domains of TNF-binding protein 1 (TBP-1) to both the alpha and beta chains of an inactive version of the heterodimeric protein hormone, human chorionic gonadotropin. The resulting molecule, termed TNF-soluble high-affinity receptor complex (SHARC), self-assembles into a heterodimeric protein containing two functional TBP-1 moieties. The TNF-SHARC is a potent inhibitor of TNF-alpha bioactivity in vitro and has a prolonged pharmacokinetic profile compared with monomeric TBP-1 in vivo. Consistent with the long half-life, the duration of action in an lipopolysaccharide-mediated proinflammatory mouse model is prolonged similarly. In a collagen-induced arthritis mouse model, this molecule demonstrates improved efficacy over monomeric TBP-1. Based on these results, we demonstrated that inactivated heterodimeric protein hormones are flexible and efficient scaffolds for the creation of soluble high-affinity receptor complexes.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/chemistry
- Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/pathology
- Cell Line, Tumor
- Cell Survival/drug effects
- Chorionic Gonadotropin/genetics
- Chorionic Gonadotropin, beta Subunit, Human/genetics
- Electrophoresis, Polyacrylamide Gel
- Female
- Glycoprotein Hormones, alpha Subunit/genetics
- Humans
- Interleukin-6/blood
- Lipopolysaccharides/pharmacology
- Mice
- Mice, Inbred C3H
- Molecular Weight
- Peptide Fragments/genetics
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/pharmacokinetics
- Recombinant Fusion Proteins/pharmacology
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Sean D McKenna
- Serono Research Institute, Rockland, Massachusetts 02370, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chai N, Gudima S, Chang J, Taylor J. Immunoadhesins containing pre-S domains of hepatitis B virus large envelope protein are secreted and inhibit virus infection. J Virol 2007; 81:4912-8. [PMID: 17329331 PMCID: PMC1900235 DOI: 10.1128/jvi.02865-06] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatitis B virus (HBV) replication produces three envelope proteins (L, M, and S) that have a common C terminus. L, the largest, contains a domain, pre-S1, not present on M. Similarly M contains a domain, pre-S2, not present on S. The pre-S1 region has important functions in the HBV life cycle. Thus, as an approach to studying these roles, the pre-S1 and/or pre-S2 sequences of HBV (serotype adw2, genotype A) were expressed as N-terminal fusions to the Fc domain of a rabbit immunoglobulin G chain. Such proteins, known as immunoadhesins (IA), were highly expressed following transfection of cultured cells and, when the pre-S1 region was present, >80% were secreted. The IA were myristoylated at a glycine penultimate to the N terminus, although mutation studies showed that this modification was not needed for secretion. As few as 30 amino acids from the N terminus of pre-S1 were both necessary and sufficient to drive secretion of IA. Even expression of pre-S1 plus pre-S2, in the absence of an immunoglobulin chain, led to efficient secretion. Overall, these studies demonstrate an unexpected ability of the N terminus of pre-S1 to promote protein secretion. In addition, some of these secreted IA, at nanomolar concentrations, inhibited infection of primary human hepatocytes either by hepatitis delta virus (HDV), a subviral agent that uses HBV envelope proteins, or HBV. These IA have potential to be part of antiviral therapies against chronic HDV and HBV, and may help understand the attachment and entry mechanisms used by these important human pathogens.
Collapse
Affiliation(s)
- Ning Chai
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111-2497, USA
| | | | | | | |
Collapse
|
32
|
Ugolini G, Marinelli S, Covaceuszach S, Cattaneo A, Pavone F. The function neutralizing anti-TrkA antibody MNAC13 reduces inflammatory and neuropathic pain. Proc Natl Acad Sci U S A 2007; 104:2985-90. [PMID: 17301229 PMCID: PMC1815293 DOI: 10.1073/pnas.0611253104] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Nerve growth factor (NGF) is involved in pain transduction mechanisms and plays a key role in many persistent pain states, notably those associated with inflammation. On this basis, both the NGF ligand and its receptor TrkA (tyrosine kinase A) represent an eligible target for pain therapy. Although the direct involvement of NGF in pain modulation is well established, the effect of a direct functional block of the TrkA receptor is still unknown. In this study, we have demonstrated that MNAC13, the only anti-TrkA monoclonal antibody for which function neutralizing properties have been clearly shown both in vitro and in vivo, induces analgesia in both inflammatory and neuropathic pain models, with a surprisingly long-lasting effect in the latter. The formalin-evoked pain licking responses are significantly reduced by the MNAC13 antibody in CD1 mice. Remarkably, treatment with the anti-TrkA antibody also produces a significant antiallodynic effect on neuropathic pain: repeated i.p. injections of MNAC13 induce significant functional recovery in mice subjected to sciatic nerve ligation, with effects persisting after administration. Furthermore, a clear synergistic effect is observed when MNAC13 is administered in combination with opioids, at doses that are not efficacious per se. This study represents a direct demonstration that neutralizing antibodies directed against the TrkA receptor may display potent analgesic effects in inflammatory and chronic pain.
Collapse
Affiliation(s)
| | - Sara Marinelli
- *Lay Line Genomics, Via di Castel Romano 100, 00128 Rome, Italy
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, Psychobiology, and Psychopharmacology, Via del Fosso di Fiorano 64, 00143 Rome, Italy; and
| | | | - Antonino Cattaneo
- *Lay Line Genomics, Via di Castel Romano 100, 00128 Rome, Italy
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Flaminia Pavone
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, Psychobiology, and Psychopharmacology, Via del Fosso di Fiorano 64, 00143 Rome, Italy; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
33
|
Shoji-Hosaka E, Kobayashi Y, Wakitani M, Uchida K, Niwa R, Nakamura K, Shitara K. Enhanced Fc-dependent cellular cytotoxicity of Fc fusion proteins derived from TNF receptor II and LFA-3 by fucose removal from Asn-linked oligosaccharides. J Biochem 2006; 140:777-83. [PMID: 17038352 DOI: 10.1093/jb/mvj207] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fucose removal from complex-type oligosaccharide of human IgGs results in a major enhancement of Fc-dependent cellular cytotoxicity. The aim of this study was to determine the effect of fucose removal on the effector function of another class of clinically important molecules that can effect cellular cytotoxicity, Fc fusion proteins. The receptors chosen for study were TNF receptor II and LFA-3, both of which have therapeutic significance. The fucosylated versions of these fusion proteins were produced in unmodified CHO cells, whereas the nonfucosylated counterparts were produced in CHO cells with alpha-1,6-fucosyltransferase, an enzyme required for fucosylation, knocked-out. Whilst binding activity of TNFRII-Fc and LFA-3-Fc were unchanged by fucose-removal, nonfucosylated Fc fusion proteins exhibited significantly higher Fc receptor gammaIIIa-binding and increased Fc-mediated cytotoxicity on target cells compared to fucosylated counterparts. Notably, in case of TNFRII-Fc, only the nonfucosylated protein exhibited potent Fc dependent cytotoxicity to transmembrane TNF-alpha expressing cells. These results prove that enhancement of Fc dependent cellular cytotoxicity by fucose-removal is effective in not only whole IgG but also Fc fusion proteins, and thus widens the potential of Fc-fusion proteins as therapeutic candidates.
Collapse
Affiliation(s)
- Emi Shoji-Hosaka
- Department of Antibody Research, Pharmaceutical Research Center, Kyowa Hakko Kogyo Co., Ltd., Machida-shi, Tokyo 194-8533
| | | | | | | | | | | | | |
Collapse
|
34
|
Qin W, Feng J, Li Y, Lin Z, Shen B. Fusion protein of CDR mimetic peptide with Fc inhibit TNF-α induced cytotoxicity. Mol Immunol 2006; 43:660-6. [PMID: 15878201 DOI: 10.1016/j.molimm.2005.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Indexed: 11/29/2022]
Abstract
The variable regions of antibodies play central roles in the binding with antigens. Based on the model of a tumour necrosis factor-alpha (TNF-alpha) neutralizing monoclonal antibody (named as Z12) with TNF-alpha, heavy chain CDR2 (HCDR2) and light chain CDR3 (LCDR3) of Z12 were found to be the most responsible to bind with TNF-alpha. A mimetic peptide (PT) was designed based on the sequence derived from HCDR2 and LCDR3. Fusion protein PT-Fc was constructed by linking PT with Fc of human IgG1 through a flexible linker (GGGGGS). The primary structural characteristics of Fc and PT-Fc were analyzed, including the flexibility, hydrophilicity and epitopes. It was demonstrated that PT and Fc in the fusion protein possessed bio-function properly and non-interfering with each other. Furthermore, PT-Fc was expressed in Escherichia coli by fusion with thioredoxin (Trx). After trx-PT-Fc was cleaved with recombinant enterokinase, PT-Fc was obtained. The results of in vitro cytotoxic assays showed that both PT and PT-Fc could efficiently inhibit TNF-alpha induced apoptosis on L929 cells. At the same micromole concentration, the inhibition activity of PT-Fc was significantly higher than PT.
Collapse
Affiliation(s)
- Weisong Qin
- Institute of Basic Medical Sciences, P.O. Box 130 (3) Taiping Road, 100850 Beijing, PR China
| | | | | | | | | |
Collapse
|
35
|
Beals JM, Shanafelt AB. Enhancing exposure of protein therapeutics. DRUG DISCOVERY TODAY. TECHNOLOGIES 2006; 3:87-94. [PMID: 24980106 DOI: 10.1016/j.ddtec.2006.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Therapeutic proteins have made a major impact on medicine, with significant expansion in the past two decades. The medicinal attributes of these agents, particularly their efficacy and often their safety profile, make protein therapeutics attractive, despite the general necessity of invasive (parenteral) delivery. This perceived hurdle has been a primary component in limiting expansion of this class of drug therapies. Strategies that reduce the frequency of administration directly provide greater convenience to the patient, and potentially greater efficacy, that can yield a significant treatment advantage.:
Collapse
Affiliation(s)
- John M Beals
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Armen B Shanafelt
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| |
Collapse
|
36
|
Meyuhas R, Noy H, Montefiori DC, Denisova G, Gershoni JM, Gross G. HIV-1 neutralization by chimeric CD4-CG10 polypeptides fused to human IgG1. Mol Immunol 2005; 42:1099-109. [PMID: 15829299 DOI: 10.1016/j.molimm.2004.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Indexed: 11/28/2022]
Abstract
The envelope glycoprotein of HIV-1 is the principal target for entry inhibitors. The use of soluble CD4 has been found to be impractical as most clinical isolates are resistant to neutralization at feasible concentrations. CG10 is one of a small group of monoclonal antibodies specific to CD4-induced epitopes, which are structurally associated with the chemokine receptor-binding site and are capable of blocking the interaction of gp120 with its obligatory co-receptor. We have reasoned that fusing the single chain Fv of CG10 with CD4 can lead to increased HIV-1 neutralization activity and that this effect could be further enhanced by engrafting this chimeric construct onto an IgG Fc. Here we report the cloning of the genes encoding the variable regions of CG10 heavy and light chains and demonstrate that when attached to human IgG1 Fc, the single chain Fv of CG10 retains the binding properties of the original mouse antibody. Fusing CG10 single chain Fv with the gp120-binding portion of CD4 on a human IgG1 Fc backbone results in stronger binding of gp120 of different tropisms and in enhanced neutralization of laboratory-adapted strains and most, but not all, clade B and clade C isolates tested. Our findings underscore the potential use of CD4-based fusion proteins in the design of HIV immuno-therapeutics.
Collapse
Affiliation(s)
- Ronit Meyuhas
- Laboratory of Immunology, MIGAL - Galilee Technology Center, P.O. Box 831, Kiryat Shmona 11016, Israel
| | | | | | | | | | | |
Collapse
|
37
|
Wang Y, Ying GX, Liu X, Wang WY, Dong JH, Ni ZM, Zhou CF. Induction of ephrin-B1 and EphB receptors during denervation-induced plasticity in the adult mouse hippocampus. Eur J Neurosci 2005; 21:2336-46. [PMID: 15932593 DOI: 10.1111/j.1460-9568.2005.04093.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Abstract It has been widely demonstrated that Eph receptors and their ephrin ligands play multiple pivotal roles in the development of the nervous system. However, less is known about their roles in the adult brain. Here we reported the expression of ephrin-B1 and its cognate EphB receptors in the adult mouse hippocampus at 3, 7, 15, 30 and 60 days after transections of the entorhinal afferents. In situ hybridization and immunohistochemistry showed the time-dependent up-regulation of ephrin-B1 in the denervated areas of the hippocampus, which initiated at 3 days postlesion (dpl), reached maximal levels at 7-15 dpl, remained slightly elevated at 30 dpl and recovered to normal levels by 60 dpl. Double labeling of ephrin-B1 and glial fibrillary acidic protein revealed that ephrin-B1-expressing cells in the denervated areas were reactive astrocytes. Furthermore, a ligand-binding assay using ephrin-B1/Fc chimera protein also displayed the up-regulation of EphB receptors in the denervated areas of the hippocampus in a similar manner to that of ephrin-B1. Within the first week postlesion, the EphB receptors were expressed by reactive astrocytes. After 7 dpl, however, EphB receptors were expressed not only by reactive astrocytes but also first by sprouting axons and later by regrowing dendrites. These results suggest that the ephrin-B1/EphB system may participate in the lesion-induced plasticity processes in the adult mouse hippocampus.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Neurobiology, Shanghai Institute of Physiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, PR China
| | | | | | | | | | | | | |
Collapse
|
38
|
Low SC, Nunes SL, Bitonti AJ, Dumont JA. Oral and pulmonary delivery of FSH–Fc fusion proteins via neonatal Fc receptor-mediated transcytosis. Hum Reprod 2005; 20:1805-13. [PMID: 15817590 DOI: 10.1093/humrep/deh896] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The alpha and beta subunits of FSH were fused to the Fc domain of IgG1 either in a single chain or a heterodimer format. These molecules were absorbed through the epithelium in lung and intestine by neonatal Fc receptor (FcRn)-mediated transcytosis. METHODS AND RESULTS Single chain and heterodimer FSH-Fc were made recombinantly in Chinese hamster ovary cells. Treatment of rats with a single s.c. dose of single chain or heterodimer FSH-Fc resulted in greater stimulation of ovarian weight (20.8+/-3.9 and 26.9+/-6.1 mg respectively) compared to those receiving vehicle (12.1+/-1.0 mg) or an equimolar dose of recombinant human FSH (14.3+/-1.7 mg). Both FSH-Fc fusion proteins were absorbed after oral dosing of newborn rats with long terminal half-lives of approximately 60 h, and pulmonary delivery in four cynomolgus monkeys produced maximum serum concentrations between 69 and 131 ng/ml with long terminal half-lives between 55 and 210 h. Serum inhibin levels increased after pulmonary dosing with single chain FSH-Fc (1.3- and 1.4-fold) and heterodimer FSH-Fc (5.9- and 7.1-fold) and remained elevated for >12 days after treatment with heterodimer FSH-Fc. CONCLUSIONS We have shown that FSH-Fc fusion proteins have increased stability in blood and improved bioactivity in vivo, and that heterodimer FSH-Fc is more active in rats and monkeys than single chain FSH-Fc. These data suggest that Fc fusion proteins offer the potential for oral and pulmonary delivery of FSH.
Collapse
Affiliation(s)
- S C Low
- Syntonix Pharmaceuticals, Inc., 9 Fourth Avenue, Waltham, MA 02451, USA.
| | | | | | | |
Collapse
|
39
|
Covaceuszach S, Cattaneo A, Lamba D. Neutralization of NGF-TrkA receptor interaction by the novel antagonistic anti-TrkA monoclonal antibody MNAC13: A structural insight. Proteins 2004; 58:717-27. [DOI: 10.1002/prot.20366] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
40
|
Qin W, Feng J, Zhang W, Li Y, Shen B. A novel TNFalpha antagonizing peptide-Fc fusion protein designed based on CDRs of TNFalpha neutralizing monoclonal antibody. Biochem Biophys Res Commun 2004; 322:1024-8. [PMID: 15336567 DOI: 10.1016/j.bbrc.2004.07.208] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Indexed: 11/27/2022]
Abstract
The variable regions of antibody molecules bind antigens with high affinity and specificity. The binding sites are imparted largely to the hypervariable portions (i.e., CDRs) of the variable region. Peptides derived from CDRs can bind antigen with similar specificity acting as mimic of antibody and become drug-designing core, although with markedly lower affinity. In order to increase the affinity and bioactivity, in this study, a novel peptide (PT) designed on CDRs of a TNFalpha neutralizing monoclonal antibody Z12 was linked with Fc fragment of human IgG1. The interaction mode of PT-linker-Fc (PLF) with TNFalpha was analyzed with computer-guided molecular modeling method. After expression in Escherichia coli and purification, recombinant PT-linker-Fc could bind directly with the TNFalpha coated on the ELISA plates. Furthermore, PLF could competitively inhibit the binding of Z12 to TNFalpha and also inhibit the TNFalpha-induced cytotoxicity on L929 cells. The TNFalpha antagonizing activity of PLF was significantly higher than that of the free peptide. This study highlights the potential of human Fc to enhance the potency of peptides designed on the CDRs of antibodies and could be useful in developing new TNFalpha antagonists.
Collapse
Affiliation(s)
- Weisong Qin
- Institute of Basic Medical Sciences, Beijing, PR China.
| | | | | | | | | |
Collapse
|
41
|
Cox GN, Smith DJ, Carlson SJ, Bendele AM, Chlipala EA, Doherty DH. Enhanced circulating half-life and hematopoietic properties of a human granulocyte colony-stimulating factor/immunoglobulin fusion protein. Exp Hematol 2004; 32:441-9. [PMID: 15145212 DOI: 10.1016/j.exphem.2004.01.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2003] [Revised: 12/31/2003] [Accepted: 01/22/2004] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The aim of this study was to determine whether fusion proteins comprising human granulocyte colony-stimulating factor (G-CSF) joined to human immunoglobulin G1 and G4 (IgG1 and IgG4) Fc and C(H) domains are biologically active and have improved pharmacokinetic and hematopoietic properties in vivo. MATERIAL AND METHODS Chimeric genes encoding human G-CSF fused to the N-termini of the Fc and C(H) domains of human IgG1 and IgG4 were constructed and used to transfect monkey COS cells. The fusion proteins were purified from the conditioned media by protein A affinity chromatography. Bioactivities of the proteins were measured in a G-CSF-dependent in vitro bioassay. Pharmacokinetic and granulopoietic properties of the G-CSF/IgG1-Fc fusion protein were measured in normal rats. RESULTS The G-CSF/IgG-Fc and G-CSF/IgG-C(H) fusion proteins were secreted from transfected COS cells primarily as disulfide-linked homodimers. On a molar basis, the purified G-CSF/IgG-Fc fusion proteins were as active as G-CSF in in vitro bioassays, whereas bioactivities of the purified G-CSF/IgG-C(H) fusion proteins were decreased 3- to 4-fold. The G-CSF/IgG1-Fc fusion protein displayed a slower plasma clearance rate and stimulated greater and longer lasting increases in circulating neutrophils and white blood cells than G-CSF following intravenous and subcutaneous administration to rats. CONCLUSION Fusion of G-CSF to human IgG domains results in homodimeric fusion proteins possessing high in vitro bioactivities, long circulating half-lives, and enhanced hematopoietic properties in vivo.
Collapse
Affiliation(s)
- George N Cox
- Bolder BioTechnology, Inc., Wheat Ridge, CO 80033, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Margotti E, Covaceuszach S, Tongiorgi E, Cattaneo A, Domenici L. TRKB signalling controls the expression of N-methyl-d-aspartate receptors in the visual cortex. Eur J Neurosci 2002; 16:1067-74. [PMID: 12383235 DOI: 10.1046/j.1460-9568.2002.02183.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
NMDA receptors (NMDARs) are multimeric proteins, the biological and functional characteristics of which depend on differential subunit assembly during postnatal development. In the present paper, we investigated whether the expression of NMDAR subunits NR1, NR2A, NR2B is influenced by neurotrophins in rat visual cortex. We used a soluble form of the TrkB receptor engineered as an immunoadhesin (TrkB-IgG) in order to block TrkB ligands. TrkB-IgG was released through a cannula implanted in the occipital pole and connected to a mini-osmotic pump. TrkB-IgG was continuously released from postnatal day 20-21 (P20-21) to P36-37. In a different group of animals used as controls, osmotic pumps were filled with saline. Different antibodies were used to stain neurons expressing NR1, NR2A and NR2B. We counted the number of neurons stained for NR2A and NR2B subunits and expressed this as percentage with respect to the total number of cresyl-violet stained neurons in each cortical layer. In the visual cortex of TrkB-IgG-treated rats, the percentage of neurons expressing NR2A was significantly increased in all cortical layers. Concerning the NR2B subunit, the percentage of stained neurons was not significantly different between TrkB-IgG-treated and control rats. The staining level for both NR2A and NR2B, but not NR1, was reduced in all cortical layers in TrkB-IgG-treated animals. In agreement with this result, the endogenous levels of NR2A and NR2B subunits were reduced in TrkB-IgG-treated animals as shown by Western blotting. Thus, TrkB signalling controls the cellular expression of NMDAR subunits in visual cortical neurons during postnatal development.
Collapse
Affiliation(s)
- Elisa Margotti
- Neuroscience Program, International School for Advanced Studies (S.I.S.S.A), via Beirut 2-4, 34014, Trieste, Italy
| | | | | | | | | |
Collapse
|
43
|
Eklund M, Axelsson L, Uhlén M, Nygren PA. Anti-idiotypic protein domains selected from protein A-based affibody libraries. Proteins 2002; 48:454-62. [PMID: 12112671 DOI: 10.1002/prot.10169] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Three pairs of small protein domains showing binding behavior in analogy with anti-idiotypic antibodies have been selected using phage display technology. From an affibody protein library constructed by combinatorial variegation of the Fc binding surface of the 58 residue staphylococcal protein A (SPA)-derived domain Z, affibody variants have been selected to the parental SPA scaffold and to two earlier identified SPA-derived affibodies. One selected affibody (Z(SPA-1)) was shown to recognize each of the five domains of wild-type SPA with dissociation constants (K(D)) in the micromolar range. The binding of the Z(SPA-1) affibody to its parental structure was shown to involve the Fc binding site of SPA, while the Fab-binding site was not involved. Similarly, affibodies showing anti-idiotypic binding characteristics were also obtained when affibodies previously selected for binding to Taq DNA polymerase and human IgA, respectively, were used as targets for selections. The potential applications for these types of affinity pairs were exemplified by one-step protein recovery using affinity chromatography employing the specific interactions between the respective protein pair members. These experiments included the purification of the Z(SPA-1) affibody from a total Escherichia coli cell lysate using protein A-Sepharose, suggesting that this protein A/antiprotein A affinity pair could provide a basis for novel affinity gene fusion systems. The use of this type of small, robust, and easily expressed anti-idiotypic affibody pair for affinity technology applications, including self-assembled protein networks, is discussed.
Collapse
Affiliation(s)
- Malin Eklund
- Department of Biotechnology, Royal Institute of Technology (KTH/SCFAB), Stockholm, Sweden
| | | | | | | |
Collapse
|
44
|
Rückert R, Brandt K, Hofmann U, Bulfone-Paus S, Paus R. IL-2-IgG2b fusion protein suppresses murine contact hypersensitivity in vivo. J Invest Dermatol 2002; 119:370-6. [PMID: 12190859 DOI: 10.1046/j.1523-1747.2002.01849.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Interleukin-15 shares several functional properties with interleukin-2, and signals through the beta and gamma chain of the interleukin-2 receptor as well as through its own high affinity alpha chain. In agreement with the concept that interleukin-2 plays a key role in type IV immune responses, we have recently shown that an IL-2-IgG2b fusion protein potently suppresses Th1-type delayed type hypersensitivity reaction and Th2-type allergic sensitization in mice. We have now compared the in vivo effects of IL-2-IgG2b fusion protein with those of IL-15-IgG2b fusion protein in a murine model of Th1-type contact hypersensitivity reaction. Daily systemic injections of IL-2-IgG2b fusion protein during the sensitization phase or application of IL-2-IgG2b fusion protein just 2 h before and 10 h after antigen challenge significantly inhibited the contact hypersensitivity ear swelling response, and this without any overt signs of associated toxicity. Even local injection of IL-2-IgG2b fusion protein into the earlobe around the time of antigen challenge inhibited the ear swelling reaction significantly. In contrast, neither systemic nor local injection of the IL-15-IgG2b fusion protein modulated the contact hypersensitivity reaction significantly. IL-2-IgG2b but not IL-15-IgG2b fusion protein reduced migration of antigen-presenting cells from the skin to local lymph nodes, inhibited the expression of CD80 and CD86, and induced a significant higher number of CD4+CD25+ T cells. Therefore, the IL-2-IgGb fusion protein offers a powerful tool for suppressing and/or preventing T-cell-mediated hypersensitivity reaction in vivo.
Collapse
Affiliation(s)
- René Rückert
- Department of Immunology and Cell Biology, Research Center Borstel, Borstel, Germany
| | | | | | | | | |
Collapse
|
45
|
Rönnmark J, Hansson M, Nguyen T, Uhlén M, Robert A, Ståhl S, Nygren PA. Construction and characterization of affibody-Fc chimeras produced in Escherichia coli. J Immunol Methods 2002; 261:199-211. [PMID: 11861078 DOI: 10.1016/s0022-1759(01)00563-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Affibody-Fc chimeras were constructed by genetic fusion between different affibody affinity proteins with prescribed specificities and an Fc fragment derived from human IgG. Using affibody ligands previously selected for binding to respiratory syncytial virus (RSV) surface protein G and Thermus aquaticus (Taq) DNA polymerase, respectively, affibody-Fc fusion proteins showing spontaneous Fc fragment-mediated homodimerization via disulfide bridges were produced in Escherichia coli and affinity purified on protein A Sepharose from bacterial periplasms at yields ranging between 1 and 6 mg/l culture. Further characterization of the chimeras using biosensor technology showed that the affibody moieties have retained high selectivities for their respective targets after fusion to the Fc fragment. Avidity effects in the target binding were observed for the affibody-Fc chimeras compared to monovalent affibody fusion proteins, indicating that both affibody moieties in the chimeras were accessible and contributed in the binding. Fusion of a head-to-tail dimeric affibody moiety to the Fc fragment resulted in tetravalent affibody constructs which showed even more pronounced avidity effects. In addition, the Fc moiety of the chimeras was demonstrated to be specifically recognized by anti-human IgG antibody enzyme conjugates. One application for this class of "artificial antibodies" was demonstrated in a western blotting experiment in which one of the anti-RSV surface protein G affibody-Fc chimeras was demonstrated to be useful for specific detection of the target protein in a complex background consisting of a total E. coli lysate. The results show that through the replacement of the Fab portion of an antibody for an alternative binding domain based on a less complicated structure, chimeric proteins compatible with bacterial production routes containing both antigen recognition domains and Fc domains can be constructed. Such "artificial antibodies" should be interesting alternatives to, for example, whole antibodies or scFv-Fc fusions as detection devices and in diagnostic or therapeutic applications.
Collapse
Affiliation(s)
- Jenny Rönnmark
- Department of Biotechnology, Royal Institute of Technology/SCFAB, SE-106 91 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
46
|
Gerlai R. Gene targeting: technical confounds and potential solutions in behavioral brain research. Behav Brain Res 2001; 125:13-21. [PMID: 11682088 DOI: 10.1016/s0166-4328(01)00282-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Gene targeting allows one to create null mutations in mice and to analyze how the mutant organism responds to the lack of a single gene product. This has facilitated the molecular dissection of such complex characteristics as mammalian brain function and behavior, including learning, memory, aggression, and maternal behavior to mention a few. However, the interpretation of the phenotypical changes that arise in null mutant mice has been questioned. The possibility that genes other than the targeted one may contribute to phenotypical alterations has been raised and the importance of compensatory mechanisms has been brought to attention. This review focuses on recent advances in the literature that illustrate the caveats associated with gene targeting and also presents an overview of potential solutions for the discussed problems.
Collapse
Affiliation(s)
- R Gerlai
- Lilly Research Laboratories, Lilly Corporate Center, Drop Code 0510, Indianapolis, IN 46285, USA.
| |
Collapse
|
47
|
Tang L, Boroughs KL, Morales T, Stedman K, Sellins K, Clarke K, McDermott M, Yang S, McCall C. Recombinant canine IL-13 receptor alpha2-Fc fusion protein inhibits canine allergen-specific-IgE production in vitro by peripheral blood mononuclear cells from allergic dogs. Vet Immunol Immunopathol 2001; 83:115-22. [PMID: 11604166 DOI: 10.1016/s0165-2427(01)00378-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Human IL-13, like IL-4, is involved in the regulation of B-cell development, IgE synthesis and allergic responses. However, because IL-13 does not affect either murine Ig class switching or IgE production in vitro, the use of murine models to study the role of IL-13 in IgE-mediated diseases has been limited. In this communication, we report that recombinant protein of canine IL-13 (rcaIL-13) stimulates production of allergen-specific-IgE in vitro by peripheral blood mononuclear cells (PBMC) from flea allergen-sensitized dogs, and that this stimulation activity is specifically inhibited by recombinant protein of canine IL-13Ralpha2 and Fc fragment of canine IgG heavy chain (rcaIL-13Ralpha2-Fc). The data suggest that the regulatory effects of IL-13 on IgE production in canine PBMC are similar to those reported in humans. Thus, canine IL-13 may be a central mediator of allergic diseases in dogs, and allergic dogs may be excellent models for research on IgE-mediated diseases in humans.
Collapse
Affiliation(s)
- L Tang
- Heska Corporation, 1613 Prospect Parkway, Fort Collins, CO 80525, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
White DM, Jensen MA, Shi X, Arnason BG. Design and expression of polymeric immunoglobulin fusion proteins: a strategy for targeting low-affinity Fcgamma receptors. Protein Expr Purif 2001; 21:446-55. [PMID: 11281720 DOI: 10.1006/prep.2001.1406] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have developed a family of cloning vectors that direct expression of fusion proteins that mimic aggregated immunoglobulin (IgG) (AIG) and immune complex function with respect to their interactions with FcgammaR and that allow for the inclusion and targeting of a second protein domain to cells expressing FcgammaR. This was accomplished by expressing multiple linear copies of the hinge and CH2 domains (HCH2) of human IgG(1) fused to the framework region of human IgG(1). Convenient restriction sites allow for the facile introduction of additional amino-terminal domains. The resulting molecule is tripartite. The carboxyl-IgG(1) framework domain provides stability and permits dimerization, and the intervening polymer provides increased effector function and targeting to FcgammaR while the amino-terminal domain can deliver an additional signal to cells expressing FcgammaR. To demonstrate the utility of the vectors, the extracellular domain of human CD8alpha was expressed as a HCH2 polymer fusion protein. The fusion proteins were secreted in useful amounts from polyclonal cell lines established in Sf9 cells following transfection and selection with G418. The biological activity of the recombinant CD8alpha-HCH2 polymers was determined and compared to those of AIG and an anti-CD16 monoclonal antibody using an in vitro assay. The activity of the fusion proteins positively correlates to the number of HCH2 units. The largest polymer tested was severalfold more potent than AIG at similar concentrations. The HCH2 polymers described here represent a new strategy in the design of recombinant proteins for the therapeutic targeting of FcgammaR in autoimmune disorders.
Collapse
Affiliation(s)
- D M White
- Department of Neurology, MC 2030, The University of Chicago, 5841 South Maryland Avenue, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
49
|
Abstract
Eph receptor tyrosine kinases are largely known for their involvement in brain development but, as some of these receptor tyrosine kinases are also expressed in adults, their possible role in the mature nervous system has begun to be explored. Evidence for the involvement of Eph receptors in synaptic plasticity, learning and memory is only emerging and needs corroboration. However, it is likely that the actions of Eph kinases in the adult brain will attract significant attention and become a fertile research area, as occurred in the case of the neurotrophins.
Collapse
Affiliation(s)
- R Gerlai
- Neuroscience Department, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285, USA.
| |
Collapse
|
50
|
Silberstein E, Dveksler G, Kaplan GG. Neutralization of hepatitis A virus (HAV) by an immunoadhesin containing the cysteine-rich region of HAV cellular receptor-1. J Virol 2001; 75:717-25. [PMID: 11134285 PMCID: PMC113968 DOI: 10.1128/jvi.75.2.717-725.2001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hepatitis A virus (HAV) infects African green monkey kidney (AGMK) cells via the HAV cellular receptor-1 (havcr-1), a mucin-like type 1 integral-membrane glycoprotein of unknown natural function. The ectodomain of havcr-1 contains an N-terminal immunoglobulin-like cysteine-rich region (D1), which binds protective monoclonal antibody (MAb) 190/4, followed by an O-glycosylated mucin-like threonine-serine-proline-rich region that extends D1 well above the cell surface. To study the interaction of HAV with havcr-1, we constructed immunoadhesins fusing the hinge and Fc portion of human IgG1 to D1 (D1-Fc) or the ectodomain of the poliovirus receptor (PVR-Fc) and expressed them in CHO cells. These immunoadhesins were secreted to the cell culture medium and purified through protein A-agarose columns. In a solid-phase assay, HAV bound to D1-Fc in a concentration-dependent manner whereas background levels of HAV bound to PVR-Fc. Binding of HAV to D1-Fc was blocked by treatment with MAb 190/4 but not with control MAb M2, which binds to a tag epitope introduced between the D1 and Fc portions of the immunoadhesin. D1-Fc neutralized approximately 1 log unit of the HAV infectivity in AGMK cells, whereas PVR-Fc had no effect in the HAV titers. A similarly poor reduction in HAV titers was observed after treating the same stock of HAV with murine neutralizing MAbs K2-4F2, K3-4C8, and VHA 813. Neutralization of poliovirus by PVR-Fc but not by D1-Fc indicated that the virus-receptor interactions were specific. These results show that D1 is sufficient for binding and neutralization of HAV and provide further evidence that havcr-1 is a functional cellular receptor for HAV.
Collapse
Affiliation(s)
- E Silberstein
- Laboratory of Hepatitis Viruses, Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|