1
|
Wani MM, Cooper JM, Migliorini M, Strickland DK. The LDL receptor related protein 1 (LRP1) facilitates ACE2-mediated endocytosis of SARS-CoV2 spike protein-containing pseudovirions. J Biol Chem 2025:110227. [PMID: 40349772 DOI: 10.1016/j.jbc.2025.110227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, employs the viral spike (S) protein to associate with host cells. While angiotensin-converting enzyme 2 (ACE2) is a major receptor for the SARS-CoV-2 spike protein, evidence reveals that other cellular receptors may also contribute to viral entry. We interrogated the role of the low-density lipoprotein receptor-related protein 1 (LRP1) in the involvement of SARS-CoV-2 viral entry. Employing surface plasmon resonance studies, we demonstrated high affinity binding of the trimeric SARS-CoV-2 spike protein to purified LRP1. Further, we observed high affinity interaction of the SARS-CoV-2 spike protein with other low-density lipoprotein receptor (LDLR) family members as well, including LRP2 and the very low-density lipoprotein receptor (VLDLR). Binding of the SARS-CoV-2 spike protein to LRP1 was mediated by its receptor binding domain (RBD). Several LRP1 ligands require surface exposed lysine residues for their interaction with LRP1, and chemical modification of lysine residues on the RBD with sulfo-NHS-acetate ablated binding to LRP1. Using cellular model systems, we demonstrated that cells expressing LRP1, but not those lacking LRP1, rapidly internalized purified 125I-labeled S1 subunit of the SARS-CoV-2 spike protein. LRP1-mediated internalization of the 125I-labeled S1 subunit was enhanced in cells expressing ACE2. By employing pseudovirion particles containing a murine leukemia virus core and luciferase reporter that express the SARS-CoV-2 spike protein on their surface, we confirmed that LRP1 facilitates ACE2-mediated psuedovirion endocytosis. Together, these data implicate LRP1, and perhaps other LDLR family members as host factors for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mashhood M Wani
- The Center for Vascular and Inflammatory Diseases, Departments of
| | - Joanna M Cooper
- The Center for Vascular and Inflammatory Diseases, Departments of; Physiology and
| | - Mary Migliorini
- The Center for Vascular and Inflammatory Diseases, Departments of
| | - Dudley K Strickland
- The Center for Vascular and Inflammatory Diseases, Departments of; Physiology and; Surgery, University of Maryland School of Medicine, Baltimore, MD 21201.
| |
Collapse
|
2
|
Reyes Ruiz A, Bhale AS, Venkataraman K, Dimitrov JD, Lacroix-Desmazes S. Binding Promiscuity of Therapeutic Factor VIII. Thromb Haemost 2025; 125:194-206. [PMID: 38950594 DOI: 10.1055/a-2358-0853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
The binding promiscuity of proteins defines their ability to indiscriminately bind multiple unrelated molecules. Binding promiscuity is implicated, at least in part, in the off-target reactivity, nonspecific biodistribution, immunogenicity, and/or short half-life of potentially efficacious protein drugs, thus affecting their clinical use. In this review, we discuss the current evidence for the binding promiscuity of factor VIII (FVIII), a protein used for the treatment of hemophilia A, which displays poor pharmacokinetics, and elevated immunogenicity. We summarize the different canonical and noncanonical interactions that FVIII may establish in the circulation and that could be responsible for its therapeutic liabilities. We also provide information suggesting that the FVIII light chain, and especially its C1 and C2 domains, could play an important role in the binding promiscuity. We believe that the knowledge accumulated over years of FVIII usage could be exploited for the development of strategies to predict protein binding promiscuity and therefore anticipate drug efficacy and toxicity. This would open a mutational space to reduce the binding promiscuity of emerging protein drugs while conserving their therapeutic potency.
Collapse
Affiliation(s)
- Alejandra Reyes Ruiz
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Aishwarya S Bhale
- Centre for Bio-Separation Technology (CBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Krishnan Venkataraman
- Centre for Bio-Separation Technology (CBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Jordan D Dimitrov
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Sébastien Lacroix-Desmazes
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| |
Collapse
|
3
|
Childers KC, Avery NG, Estrada Alamo KA, Davulcu O, Haynes RM, Lollar P, Doering CB, Coxon CH, Spiegel PC. Structure of coagulation factor VIII bound to a patient-derived anti-C1 domain antibody inhibitor. Blood 2023; 142:197-201. [PMID: 37192299 PMCID: PMC10352601 DOI: 10.1182/blood.2023020181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/07/2023] [Accepted: 04/25/2023] [Indexed: 05/18/2023] Open
Abstract
The development of pathogenic antibody inhibitors against coagulation factor VIII (FVIII) occurs in ∼30% of patients with congenital hemophilia A receiving FVIII replacement therapy, as well as in all cases of acquired hemophilia A. KM33 is an anti-C1 domain antibody inhibitor previously isolated from a patient with severe hemophilia A. In addition to potently blocking FVIII binding to von Willebrand factor and phospholipid surfaces, KM33 disrupts FVIII binding to lipoprotein receptor-related protein 1 (LRP1), which drives FVIII hepatic clearance and antigen presentation in dendritic cells. Here, we report on the structure of FVIII bound to NB33, a recombinant derivative of KM33, via single-particle cryo-electron microscopy. Structural analysis revealed that the NB33 epitope localizes to the FVIII residues R2090-S2094 and I2158-R2159, which constitute membrane-binding loops in the C1 domain. Further analysis revealed that multiple FVIII lysine and arginine residues, previously shown to mediate binding to LRP1, dock onto an acidic cleft at the NB33 variable domain interface, thus blocking a putative LRP1 binding site. Together, these results demonstrate a novel mechanism of FVIII inhibition by a patient-derived antibody inhibitor and provide structural evidence for engineering FVIII with reduced LRP1-mediated clearance.
Collapse
Affiliation(s)
| | - Nathan G. Avery
- Chemistry Department, Western Washington University, Bellingham, WA
| | | | - Omar Davulcu
- Pacific Northwest Center for Cryo-EM, Oregon Health & Science University, Portland, OR
- Pacific Northwest National Laboratory, Environmental Molecular Sciences Laboratory, Richland, WA
| | - Rose Marie Haynes
- Pacific Northwest Center for Cryo-EM, Oregon Health & Science University, Portland, OR
- Pacific Northwest National Laboratory, Environmental Molecular Sciences Laboratory, Richland, WA
| | - Pete Lollar
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA
| | - Christopher B. Doering
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA
- Expression Therapeutics Inc, Tucker, GA
| | - Carmen H. Coxon
- National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, United Kingdom
| | - P. Clint Spiegel
- Chemistry Department, Western Washington University, Bellingham, WA
| |
Collapse
|
4
|
Camilleri E, van Rein N, van Vlijmen BJM, Biedermann JS, Kruip MJHA, Leebeek FW, van der Meer FJ, Cobbaert CM, Cannegieter SC, Lijfering WM. Influence of rosuvastatin on apolipoproteins and coagulation factor levels: Results from the STAtin Reduce Thrombophilia trial. Res Pract Thromb Haemost 2023; 7:100063. [PMID: 36923709 PMCID: PMC10009537 DOI: 10.1016/j.rpth.2023.100063] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 02/05/2023] Open
Abstract
Background The STAtins Reduce Thrombophilia trial showed that, in patients with prior venous thrombosis, rosuvastatin decreased various coagulation factor levels. Objectives Here, we investigated the hypothesis that statins decrease coagulation factor levels through shared mechanisms of synthesis or regulatory pathways with apolipoproteins. Methods We measured the levels of apolipoprotein (Apo)A-I, A-II, A-IV, (a), B-100, B-total, C-I, C-II, C-III, and E in patients (n = 126) randomized to 28 days of rosuvastatin use. We assessed the association between apolipoproteins and coagulation factors at baseline using linear regression. The mean difference in apolipoprotein levels between baseline and after 28 days of rosuvastatin use was determined through linear regression, adjusting for age, sex, and body mass index. Coagulation factors were added to this model to determine if the lowering of apolipoproteins by rosuvastatin was linked with coagulation factor levels. Results At baseline, levels of all apolipoproteins, except Apo(a), were positively associated with FVII, FIX, and FXI. Apolipoproteins levels, except for ApoA-I, A-IV, and Apo(a), were decreased after 28 days of rosuvastatin. ApoB-100 showed the largest mean decrease of -0.43 g/L (95% CI = -0.46 to -0.40). The decrease in ApoC-I and C-III levels was associated with a decrease in FVII, whereas the decrease in apoA-II, B-100, and B-total was associated with a decrease in FXI. The decrease in apolipoproteins was neither associated with FVIII or vWF decrease nor with endogenous thrombin potential changes. Conclusions Rosuvastatin decreases the level of several apolipoproteins, but this decrease was associated only with a decrease in FVII and XI and not with FVIII/vWF.
Collapse
Affiliation(s)
- Eleonora Camilleri
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nienke van Rein
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Pharmacy, Leiden University Medical Center, Leiden, the Netherlands
| | - Bart J M van Vlijmen
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Joseph S Biedermann
- Department of Hematology, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marieke J H A Kruip
- Department of Hematology, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands.,Thrombosis Service Star-shl, Rotterdam, the Netherlands
| | - Frank W Leebeek
- Department of Hematology, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Felix J van der Meer
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Suzanne C Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Willem M Lijfering
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
5
|
Chun H, Kurasawa JH, Olivares P, Marakasova ES, Shestopal SA, Hassink GU, Karnaukhova E, Migliorini M, Obi JO, Smith AK, Wintrode PL, Durai P, Park K, Deredge D, Strickland DK, Sarafanov AG. Characterization of interaction between blood coagulation factor VIII and LRP1 suggests dynamic binding by alternating complex contacts. J Thromb Haemost 2022; 20:2255-2269. [PMID: 35810466 PMCID: PMC9804390 DOI: 10.1111/jth.15817] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/15/2022] [Accepted: 07/01/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Deficiency in blood coagulation factor VIII (FVIII) results in life-threating bleeding (hemophilia A) treated by infusions of FVIII concentrates. To improve disease treatment, FVIII has been modified to increase its plasma half-life, which requires understanding mechanisms of FVIII catabolism. An important catabolic actor is hepatic low density lipoprotein receptor-related protein 1 (LRP1), which also regulates many other clinically significant processes. Previous studies showed complexity of FVIII site for binding LRP1. OBJECTIVES To characterize binding sites between FVIII and LRP1 and suggest a model of the interaction. METHODS A series of recombinant ligand-binding complement-type repeat (CR) fragments of LRP1 including mutated variants was generated in a baculovirus system and tested for FVIII interaction using surface plasmon resonance, tissue culture model, hydrogen-deuterium exchange mass spectrometry, and in silico. RESULTS Multiple CR doublets within LRP1 clusters II and IV were identified as alternative FVIII-binding sites. These interactions follow the canonical binding mode providing major binding energy, and additional weak interactions are contributed by adjacent CR domains. A representative CR doublet was shown to have multiple contact sites on FVIII. CONCLUSIONS FVIII and LRP1 interact via formation of multiple complex contacts involving both canonical and non-canonical binding combinations. We propose that FVIII-LRP1 interaction occurs via switching such alternative binding combinations in a dynamic mode, and that this mechanism is relevant to other ligand interactions of the low-density lipoprotein receptor family members including LRP1.
Collapse
Affiliation(s)
- Haarin Chun
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - James H. Kurasawa
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Present address:
Biologics Engineering, R&D, AstraZeneca, GaithersburgMarylandUSA
| | - Philip Olivares
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Ekaterina S. Marakasova
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Present address:
(1) Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver SpringMarylandUSA
- Present address:
George Mason University, School of Systems Biology, FairfaxVirginiaUSA
| | - Svetlana A. Shestopal
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Gabriela U. Hassink
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Present address:
GSK‐Rockville Center for Vaccines Research, RockvilleMarylandUSA
| | - Elena Karnaukhova
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Mary Migliorini
- Center for Vascular and Inflammatory DiseasesDepartments of Surgery and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Juliet O. Obi
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Ally K. Smith
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Patrick L. Wintrode
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Prasannavenkatesh Durai
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Keunwan Park
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Daniel Deredge
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory DiseasesDepartments of Surgery and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Andrey G. Sarafanov
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
6
|
Cadé M, Muñoz-Garcia J, Babuty A, Fouassier M, Heymann MF, Monahan PE, Heymann D. FVIII at the crossroad of coagulation, bone and immune biology: Emerging evidence of biological activities beyond hemostasis. Drug Discov Today 2021; 27:102-116. [PMID: 34311113 DOI: 10.1016/j.drudis.2021.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/27/2021] [Accepted: 07/19/2021] [Indexed: 12/19/2022]
Abstract
Hemophilia A is an X-linked hereditary disorder that results from deficient coagulation factor VIII (FVIII) activity, leading to spontaneous bleeding episodes, particularly in joints and muscles. FVIII deficiency has been associated with altered bone remodeling, dysregulated macrophage polarization, and inflammatory processes that are associated with the neoformation of abnormal blood vessels. Treatment based on FVIII replacement can lead to the development of inhibitors that render FVIII concentrate infusion ineffective. In this context, hemophilia has entered a new therapeutic era with the development of new drugs, such as emicizumab, that seek to restore the hemostatic balance by bypassing pathologically acquired antibodies. We discuss the potential extrahemostatic functions of FVIII that may be crucial for defining future therapies in hemophilia.
Collapse
Affiliation(s)
- Marie Cadé
- Université de Nantes, INSERM, Institut de Cancérologie de l'Ouest, Saint-Herblain 44805, France
| | - Javier Muñoz-Garcia
- Université de Nantes, INSERM, Institut de Cancérologie de l'Ouest, Saint-Herblain 44805, France
| | - Antoine Babuty
- Université de Nantes, INSERM, Institut de Cancérologie de l'Ouest, Saint-Herblain 44805, France; Department of Haemostasis, CHU de Nantes, France
| | | | - Marie-Francoise Heymann
- Université de Nantes, INSERM, Institut de Cancérologie de l'Ouest, Saint-Herblain 44805, France
| | - Paul E Monahan
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Dominique Heymann
- Université de Nantes, INSERM, Institut de Cancérologie de l'Ouest, Saint-Herblain 44805, France; University of Sheffield, Department of Oncology and Metabolism, Sheffield, UK.
| |
Collapse
|
7
|
Marakasova E, Olivares P, Karnaukhova E, Chun H, Hernandez NE, Kurasawa JH, Hassink GU, Shestopal SA, Strickland DK, Sarafanov AG. Molecular chaperone RAP interacts with LRP1 in a dynamic bivalent mode and enhances folding of ligand-binding regions of other LDLR family receptors. J Biol Chem 2021; 297:100842. [PMID: 34058195 PMCID: PMC8239462 DOI: 10.1016/j.jbc.2021.100842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
The low-density lipoprotein receptor (LDLR) family of receptors are cell-surface receptors that internalize numerous ligands and play crucial role in various processes, such as lipoprotein metabolism, hemostasis, fetal development, etc. Previously, receptor-associated protein (RAP) was described as a molecular chaperone for LDLR-related protein 1 (LRP1), a prominent member of the LDLR family. We aimed to verify this role of RAP for LRP1 and two other LDLR family receptors, LDLR and vLDLR, and to investigate the mechanisms of respective interactions using a cell culture model system, purified system, and in silico modelling. Upon coexpression of RAP with clusters of the ligand-binding complement repeats (CRs) of the receptors in secreted form in insect cells culture, the isolated proteins had increased yield, enhanced folding, and improved binding properties compared with proteins expressed without RAP, as determined by circular dichroism and surface plasmon resonance. Within LRP1 CR-clusters II and IV, we identified multiple sites comprised of adjacent CR doublets, which provide alternative bivalent binding combinations with specific pairs of lysines on RAP. Mutational analysis of these lysines within each of isolated RAP D1/D2 and D3 domains having high affinity to LRP1 and of conserved tryptophans on selected CR-doublets of LRP1, as well as in silico docking of a model LRP1 CR-triplet with RAP, indicated a universal role for these residues in interaction of RAP and LRP1. Consequently, we propose a new model of RAP interaction with LDLR family receptors based on switching of the bivalent contacts between molecules over time in a dynamic mode.
Collapse
Affiliation(s)
- Ekaterina Marakasova
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Philip Olivares
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Elena Karnaukhova
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Haarin Chun
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Nancy E Hernandez
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - James H Kurasawa
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gabriela U Hassink
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Svetlana A Shestopal
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrey G Sarafanov
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA.
| |
Collapse
|
8
|
Cooper JM, Lathuiliere A, Migliorini M, Arai AL, Wani MM, Dujardin S, Muratoglu SC, Hyman BT, Strickland DK. Regulation of tau internalization, degradation, and seeding by LRP1 reveals multiple pathways for tau catabolism. J Biol Chem 2021; 296:100715. [PMID: 33930462 PMCID: PMC8164048 DOI: 10.1016/j.jbc.2021.100715] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
In Alzheimer's disease (AD), pathological forms of tau are transferred from cell to cell and “seed” aggregation of cytoplasmic tau. Phosphorylation of tau plays a key role in neurodegenerative tauopathies. In addition, apolipoprotein E (apoE), a major component of lipoproteins in the brain, is a genetic risk determinant for AD. The identification of the apoE receptor, low-density lipoprotein receptor–related protein 1 (LRP1), as an endocytic receptor for tau raises several questions about the role of LRP1 in tauopathies: is internalized tau, like other LRP1 ligands, delivered to lysosomes for degradation, and does LRP1 internalize pathological tau leading to cytosolic seeding? We found that LRP1 rapidly internalizes 125I-labeled tau, which is then efficiently degraded in lysosomal compartments. Surface plasmon resonance experiments confirm high affinity binding of tau and the tau microtubule-binding domain to LRP1. Interestingly, phosphorylated forms of recombinant tau bind weakly to LRP1 and are less efficiently internalized by LRP1. LRP1-mediated uptake of tau is inhibited by apoE, with the apoE4 isoform being the most potent inhibitor, likely because of its higher affinity for LRP1. Employing post-translationally–modified tau derived from brain lysates of human AD brain tissue, we found that LRP1-expressing cells, but not LRP1-deficient cells, promote cytosolic tau seeding in a process enhanced by apoE. These studies identify LRP1 as an endocytic receptor that binds and processes monomeric forms of tau leading to its degradation and promotes seeding by pathological forms of tau. The balance of these processes may be fundamental to the spread of neuropathology across the brain in AD.
Collapse
Affiliation(s)
- Joanna M Cooper
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Aurelien Lathuiliere
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Mary Migliorini
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Allison L Arai
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mashhood M Wani
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Simon Dujardin
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Selen C Muratoglu
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bradley T Hyman
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA.
| | - Dudley K Strickland
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
9
|
Chun H, Pettersson JR, Shestopal SA, Wu WW, Marakasova ES, Olivares P, Surov SS, Ovanesov MV, Shen RF, Sarafanov AG. Characterization of protein unable to bind von Willebrand factor in recombinant factor VIII products. J Thromb Haemost 2021; 19:954-966. [PMID: 33527662 DOI: 10.1111/jth.15257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Therapeutic products with coagulation factor VIII (FVIII) have a wide range of specific activities, implying presence of protein with altered structure. Previous studies showed that recombinant FVIII products (rFVIII) contain a fraction (FVIIIFT ) unable to bind von Willebrand factor (VWF) and reported to lack activity. Because of loss of function(s), FVIIIFT can be defined as a product-related impurity, whose properties and levels in rFVIII products should be investigated. OBJECTIVE To isolate and characterize the FVIIIFT fraction in rFVIII products. METHODS Protein fractions unable (FVIIIFT ) and able (FVIIIEL ) to bind VWF were isolated from rFVIII products using immobilized VWF affinity chromatography (IVAC) and characterized by gel electrophoresis, immunoblotting, FVIII activity test, surface plasmon resonance, mass spectrometry, and for plasma clearance in mice. RESULTS AND CONCLUSIONS A robust IVAC methodology was developed and applied for analysis of 10 rFVIII products marketed in the United States. FVIIIFT was found at various contents (0.4%-21.5%) in all products. Compared with FVIIIEL , FVIIIFT had similar patterns of polypeptide bands by gel electrophoresis, but lower functional activity. In several representative products, FVIIIFT was found to have reduced sulfation at Tyr1680, important for VWF binding, decreased interaction with a low-density lipoprotein receptor-related protein 1 fragment, and faster plasma clearance in mice. These findings provide basic characterization of FVIIIFT and demonstrate a potential for IVAC to control this impurity in rFVIII products to improve their efficacy in therapy of hemophilia A.
Collapse
Affiliation(s)
- Haarin Chun
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - John R Pettersson
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Svetlana A Shestopal
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Wells W Wu
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Ekaterina S Marakasova
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Philip Olivares
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Stepan S Surov
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Mikhail V Ovanesov
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Rong-Fong Shen
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| | - Andrey G Sarafanov
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
10
|
Fouët G, Gout E, Wicker-Planquart C, Bally I, De Nardis C, Dedieu S, Chouquet A, Gaboriaud C, Thielens NM, Kleman JP, Rossi V. Complement C1q Interacts With LRP1 Clusters II and IV Through a Site Close but Different From the Binding Site of Its C1r and C1s-Associated Proteases. Front Immunol 2020; 11:583754. [PMID: 33193398 PMCID: PMC7609443 DOI: 10.3389/fimmu.2020.583754] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
LRP1 is a large endocytic modular receptor that plays a crucial role in the scavenging of apoptotic material through binding to pattern-recognition molecules. It is a membrane anchored receptor of the LDL receptor family with 4 extracellular clusters of ligand binding modules called cysteine rich complement-type repeats that are involved in the interaction of LRP1 with its numerous ligands. Complement C1q was shown to interact with LRP1 and to be implicated in the phagocytosis of apoptotic cells. The present work aimed at exploring how these two large molecules interact at the molecular level using a dissection strategy. For that purpose, recombinant LRP1 clusters II, III and IV were produced in mammalian HEK293F cells and their binding properties were investigated. Clusters II and IV were found to interact specifically and efficiently with C1q with KDs in the nanomolar range. The use of truncated C1q fragments and recombinant mutated C1q allowed to localize more precisely the binding site for LRP1 on the collagen-like regions of C1q (CLRs), nearby the site that is implicated in the interaction with the cognate protease tetramer C1r2s2. This site could be a common anchorage for other ligands of C1q CLRs such as sulfated proteoglycans and Complement receptor type 1. The use of a cellular model, consisting in CHO LRP1-null cells transfected with full-length LRP1 or a cluster IV minireceptor (mini IV) confirmed that mini IV interacts with C1q at the cell membrane as well as full-length LRP1. Further cellular interaction studies finally highlighted that mini IV can endorse the full-length LRP1 binding efficiency for apoptotic cells and that C1q has no impact on this interaction.
Collapse
Affiliation(s)
| | - Evelyne Gout
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | | | - Isabelle Bally
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Camilla De Nardis
- Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Stéphane Dedieu
- Université de Reims Champagne-Ardenne, UMR CNRS 7369 MEDyC, Reims, France
| | - Anne Chouquet
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | | | | | | | | |
Collapse
|
11
|
Arai AL, Migliorini M, Au DT, Hahn-Dantona E, Peeney D, Stetler-Stevenson WG, Muratoglu SC, Strickland DK. High-Affinity Binding of LDL Receptor-Related Protein 1 to Matrix Metalloprotease 1 Requires Protease:Inhibitor Complex Formation. Biochemistry 2020; 59:2922-2933. [PMID: 32702237 DOI: 10.1021/acs.biochem.0c00442] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Matrix metalloprotease (MMP) activation contributes to the degradation of the extracellular matrix (ECM), resulting in a multitude of pathologies. Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifaceted endocytic and signaling receptor that is responsible for internalization and lysosomal degradation of diverse proteases, protease inhibitors, and lipoproteins along with numerous other proteins. In this study, we identified MMP-1 as a novel LRP1 ligand. Binding studies employing surface plasmon resonance revealed that both proMMP-1 and active MMP-1 bind to purified LRP1 with equilibrium dissociation constants (KD) of 19 and 25 nM, respectively. We observed that human aortic smooth muscle cells readily internalize and degrade 125I-labeled proMMP-1 in an LRP1-mediated process. Our binding data also revealed that all tissue inhibitors of metalloproteases (TIMPs) bind to LRP1 with KD values ranging from 23 to 33 nM. Interestingly, the MMP-1/TIMP-1 complex bound to LRP1 with an affinity (KD = 0.6 nM) that was 30-fold higher than that of either component alone, revealing that LRP1 prefers the protease:inhibitor complex as a ligand. Of note, modification of lysine residues on either proMMP-1 or TIMP-1 ablated the ability of the MMP-1/TIMP-1 complex to bind to LRP1. LRP1's preferential binding to enzyme:inhibitor complexes was further supported by the higher binding affinity for proMMP-9/TIMP-1 complexes than for either of these two components alone. LRP1 has four clusters of ligand-binding repeats, and MMP-1, TIMP-1, and MMP-1/TIMP-1 complexes bound to cluster III most avidly. Our results reveal an important role for LRP1 in controlling ECM homeostasis by regulating MMP-1 and MMP-9 levels.
Collapse
Affiliation(s)
| | | | | | | | - David Peeney
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - William G Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | | | | |
Collapse
|
12
|
Falach R, Sapoznikov A, Gal Y, Elhanany E, Evgy Y, Shifman O, Aftalion M, Ehrlich S, Lazar S, Sabo T, Kronman C, Mazor O. The low density receptor-related protein 1 plays a significant role in ricin-mediated intoxication of lung cells. Sci Rep 2020; 10:9007. [PMID: 32488096 PMCID: PMC7265403 DOI: 10.1038/s41598-020-65982-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/11/2020] [Indexed: 01/15/2023] Open
Abstract
Ricin, a highly lethal plant-derived toxin, is a potential biological threat agent due to its high availability, ease of production and the lack of approved medical countermeasures for post-exposure treatment. To date, no specific ricin receptors were identified. Here we show for the first time, that the low density lipoprotein receptor-related protein-1 (LRP1) is a major target molecule for binding of ricin. Pretreating HEK293 acetylcholinesterase-producer cells with either anti-LRP1 antibodies or with Receptor-Associated Protein (a natural LRP1 antagonist), or using siRNA to knock-down LRP1 expression resulted in a marked reduction in their sensitivity towards ricin. Binding assays further demonstrated that ricin bound exclusively to the cluster II binding domain of LRP1, via the ricin B subunit. Ricin binding to the cluster II binding domain of LRP1 was significantly reduced by an anti-ricin monoclonal antibody, which confers high-level protection to ricin pulmonary-exposed mice. Finally, we tested the contribution of LRP1 receptor to ricin intoxication of lung cells derived from mice. Treating these cells with anti-LRP1 antibody prior to ricin exposure, prevented their intoxication. Taken together, our findings clearly demonstrate that the LRP1 receptor plays an important role in ricin-induced pulmonary intoxications.
Collapse
Affiliation(s)
- Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel.
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Eytan Elhanany
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Yentl Evgy
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Ohad Shifman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Moshe Aftalion
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Sharon Ehrlich
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Shlomi Lazar
- Department of Pharmacology, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| | - Ohad Mazor
- Department of Infectious Diseases, Israel Institute for Biological Research, 19 Reuven Lerer St., Ness-Ziona, 76100, Israel
| |
Collapse
|
13
|
Migliorini M, Li SH, Zhou A, Emal CD, Lawrence DA, Strickland DK. High-affinity binding of plasminogen-activator inhibitor 1 complexes to LDL receptor-related protein 1 requires lysines 80, 88, and 207. J Biol Chem 2020; 295:212-222. [PMID: 31792055 PMCID: PMC6952620 DOI: 10.1074/jbc.ra119.010449] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/25/2019] [Indexed: 11/06/2022] Open
Abstract
It is well-established that complexes of plasminogen-activator inhibitor 1 (PAI-1) with its target enzymes bind tightly to low-density lipoprotein (LDL) receptor-related protein 1 (LRP1), but the molecular details of this interaction are not well-defined. Furthermore, considerable controversy exists in the literature regarding the nature of the interaction of free PAI-1 with LRP1. In this study, we examined the binding of free PAI-1 and complexes of PAI-1 with low-molecular-weight urokinase-type plasminogen activator to LRP1. Our results confirmed that uPA:PAI-1 complexes bind LRP1 with ∼100-fold increased affinity over PAI-1 alone. Chemical modification of PAI-1 confirmed an essential requirement of lysine residues in PAI-1 for the interactions of both PAI-1 and uPA:PAI-1 complexes with LRP1. Results of surface plasmon resonance measurements supported a bivalent binding model in which multiple sites on PAI-1 and uPA:PAI-1 complexes interact with complementary sites on LRP1. An ionic-strength dependence of binding suggested the critical involvement of two charged residues for the interaction of PAI-1 with LRP1 and three charged residues for the interaction of uPA:PAI-1 complexes with LRP1. An enhanced affinity resulting from the interaction of three regions of the uPA:PAI-1 complex with LDLa repeats on LRP1 provided an explanation for the increased affinity of uPA:PAI-1 complexes for LRP1. Mutational analysis revealed an overlap between LRP1 binding and binding of a small-molecule inhibitor of PAI-1, CDE-096, confirming an important role for Lys-207 in the interaction of PAI-1 with LRP1 and of the orientations of Lys-207, -88, and -80 for the interaction of uPA:PAI-1 complexes with LRP1.
Collapse
Affiliation(s)
- Mary Migliorini
- Center for Vascular and Inflammatory Diseases and the Departments of Surgery and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Shih-Hon Li
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Anqi Zhou
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Cory D Emal
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197
| | - Daniel A Lawrence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109.
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases and the Departments of Surgery and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201.
| |
Collapse
|
14
|
Kis-Toth K, Rajani GM, Simpson A, Henry KL, Dumont J, Peters RT, Salas J, Loh C. Recombinant factor VIII Fc fusion protein drives regulatory macrophage polarization. Blood Adv 2018; 2:2904-2916. [PMID: 30396910 PMCID: PMC6234359 DOI: 10.1182/bloodadvances.2018024497] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
The main complication of replacement therapy with factor in hemophilia A (HemA) is the formation of inhibitors (neutralizing anti-factor VIII [FVIII] antibodies) in ∼30% of severe HemA patients. Because these inhibitors render replacement FVIII treatment essentially ineffective, preventing or eliminating them is of top priority in disease management. The extended half-life recombinant FVIII Fc fusion protein (rFVIIIFc) is an approved therapy for HemA patients. In addition, it has been reported that rFVIIIFc may induce tolerance to FVIII more readily than FVIII alone in HemA patients that have developed inhibitors. Given that the immunoglobulin G1 Fc region has the potential to interact with immune cells expressing Fc receptors (FcRs) and thereby affect the immune response to rFVIII, we investigated how human macrophages, expressing both FcRs and receptors reported to bind FVIII, respond to rFVIIIFc. We show herein that rFVIIIFc, but not rFVIII, uniquely skews macrophages toward an alternatively activated regulatory phenotype. rFVIIIFc initiates signaling events that result in morphological changes, as well as a specific gene expression and metabolic profile that is characteristic of the regulatory type Mox/M2-like macrophages. Further, these changes are dependent on rFVIIIFc-FcR interactions. Our findings elucidate mechanisms of potential immunomodulatory properties of rFVIIIFc.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joe Salas
- Bioverativ, a Sanofi company, Waltham, MA; and
| | | |
Collapse
|
15
|
Shestopal SA, Hao JJ, Karnaukhova E, Liang Y, Ovanesov MV, Lin M, Kurasawa JH, Lee TK, Mcvey JH, Sarafanov AG. Expression and characterization of a codon-optimized blood coagulation factor VIII. J Thromb Haemost 2017; 15:709-720. [PMID: 28109042 DOI: 10.1111/jth.13632] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Indexed: 08/31/2023]
Abstract
Essentials Recombinant factor VIII (FVIII) is known to be expressed at a low level in cell culture. To increase expression, we used codon-optimization of a B-domain deleted FVIII (BDD-FVIII). This resulted in 7-fold increase of the expression level in cell culture. The biochemical properties of codon-optimized BDD-FVIII were similar to the wild-type protein. SUMMARY Background Production of recombinant factor VIII (FVIII) is challenging because of its low expression. It was previously shown that codon-optimization of a B-domain-deleted FVIII (BDD-FVIII) cDNA resulted in increased protein expression. However, it is well recognized that synonymous mutations may affect the protein structure and function. Objectives To compare biochemical properties of a BDD-FVIII variants expressed from codon-optimized and wild-type cDNAs (CO and WT, respectively). Methods Each variant of the BDD-FVIII was expressed in several independent Chinese hamster ovary (CHO) cell lines, generated using a lentiviral platform. The proteins were purified by two-step affinity chromatography and analyzed in parallel by PAGE-western blot, mass spectrometry, circular dichroism, surface plasmon resonance, and chromogenic, clotting and thrombin generation assays. Results and conclusion The average yield of the CO was 7-fold higher than WT, whereas both proteins were identical in the amino acid sequences (99% coverage) and very similar in patterns of the molecular fragments (before and after thrombin cleavage), glycosylation and tyrosine sulfation, secondary structures and binding to von Willebrand factor and to a fragment of the low-density lipoprotein receptor-related protein 1. The CO preparations had on average 1.5-fold higher FVIII specific activity (activity normalized to protein mass) than WT preparations, which was attributed to better preservation of the CO structure as a result of considerably higher protein concentrations during the production. We concluded that the codon-optimization of the BDD-FVIII resulted in significant increase of its expression and did not affect the structure-function properties.
Collapse
Affiliation(s)
- S A Shestopal
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - J-J Hao
- Poochon Scientific, Frederick, MD, USA
| | - E Karnaukhova
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Y Liang
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - M V Ovanesov
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - M Lin
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - J H Kurasawa
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - T K Lee
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - J H Mcvey
- School of Biosciences and Medicine, University of Surrey, Surrey, UK
| | - A G Sarafanov
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|