Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
J Cereb Blood Flow Metab ; 38(4): 727-740, 2018 04.
Article in English | MEDLINE | ID: mdl-29140158

ABSTRACT

Current methods for examining antibody trafficking are either non-quantitative such as immunocytochemistry or require antibody labeling with tracers. We have developed a multiplexed quantitative method for antibody 'tracking' in endosomal compartments of brain endothelial cells. Rat brain endothelial cells were co-incubated with blood-brain barrier (BBB)-crossing FC5, monovalent FC5Fc or bivalent FC5Fc fusion antibodies and control antibodies. Endosomes were separated using sucrose-density gradient ultracentrifugation and analyzed using multiplexed mass spectrometry to simultaneously quantify endosomal markers, receptor-mediated transcytosis (RMT) receptors and the co-incubated antibodies in each fraction. The quantitation showed that markers of early endosomes were enriched in high-density fractions (HDF), whereas markers of late endosomes and lysosomes were enriched in low-density fractions (LDF). RMT receptors, including transferrin receptor, showed a profile similar to that of early endosome markers. The in vitro BBB transcytosis rates of antibodies were directly proportional to their partition into early endosome fractions of brain endothelial cells. Addition of the Fc domain resulted in facilitated antibody 'redistribution' from LDF into HDF and additionally into multivesicular bodies (MVB). Sorting of various FC5 antibody formats away from late endosomes and lysosomes and into early endosomes and a subset of MVB results in increased antibody transcytosis at the abluminal side of the BBB.


Subject(s)
Antibodies/metabolism , Blood-Brain Barrier/physiology , Endosomes/physiology , Transcytosis/physiology , Animals , Antibodies/cerebrospinal fluid , Antigens, CD , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Lysosomes/metabolism , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Receptors, Transferrin
2.
Gene ; 576(1 Pt 2): 319-32, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26497270

ABSTRACT

Muscle atrophy results from a range of physiological conditions, including immobilization, spinal cord damage, inflammation and aging. In this study we describe two genes, NEFA-interacting nuclear protein 30 (Nip30) and RING Finger and SPRY domain containing 1 (Rspry1), which have not previously been characterized or shown to be expressed in skeletal muscle. Furthermore, Nip30 and Rspry1 were transcriptionally induced in response to neurogenic muscle wasting in mice and were also found to be expressed endogenously at the RNA and protein level in C2C12 mouse muscle cells. Interestingly, during analysis of Nip30 and Rspry1 it was observed that these genes share a 230 base pair common regulatory region that contains several putative transcription regulatory elements. In order to assess the transcriptional activity of the Nip30 and Rspry1 regulatory regions, a fragment of the promoter of each gene was cloned, fused to a reporter gene, and transfected into cells. The Nip30 and Rspry1 reporters were both found to have significant transcriptional activity in cultured cells. Furthermore, the Nip30-Rspry1 common regulatory region contains a conserved E-box enhancer, which is an element bound by myogenic regulatory factors that function in the regulation of muscle-specific gene expression. Therefore, in order to determine if the predicted E-box was functional, Nip30 and Rspry1 reporters were transfected into cells ectopically expressing the myogenic regulatory factor, MyoD1, resulting in significant induction of both reporter genes. In addition, mutation of the conserved E-box element eliminated MyoD1 activation of the Nip30 and Rspry1 reporters. Finally, GFP-tagged Nip30 was found to localize to the nucleus, while GFP-tagged Rspry1 was found to localize to the cytoplasm of muscle cells.


Subject(s)
DNA-Binding Proteins/genetics , Muscle, Skeletal/physiology , Nuclear Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Conserved Sequence , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , E-Box Elements , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscular Atrophy/genetics , Muscular Atrophy/physiopathology , MyoD Protein/genetics , MyoD Protein/metabolism , Nuclear Proteins/metabolism , Protein Structure, Tertiary
3.
FASEB J ; 28(11): 4764-78, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25070367

ABSTRACT

The blood-brain barrier (BBB) prevents the access of therapeutic antibodies to central nervous system (CNS) targets. The engineering of bispecific antibodies in which a therapeutic "arm" is combined with a BBB-transcytosing arm can significantly enhance their brain delivery. The BBB-permeable single-domain antibody FC5 was previously isolated by phenotypic panning of a naive llama single-domain antibody phage display library. In this study, FC5 was engineered as a mono- and bivalent fusion with the human Fc domain to optimize it as a modular brain delivery platform. In vitro studies demonstrated that the bivalent fusion of FC5 with Fc increased the rate of transcytosis (Papp) across brain endothelial monolayer by 25% compared with monovalent fusion. Up to a 30-fold enhanced apparent brain exposure (derived from serum and cerebrospinal fluid pharmacokinetic profiles) of FC5- compared with control domain antibody-Fc fusions after systemic dosing in rats was observed. Systemic pharmacological potency was evaluated in the Hargreaves model of inflammatory pain using the BBB-impermeable neuropeptides dalargin and neuropeptide Y chemically conjugated with FC5-Fc fusion proteins. Improved serum pharmacokinetics of Fc-fused FC5 contributed to a 60-fold increase in pharmacological potency compared with the single-domain version of FC5; bivalent and monovalent FC5 fusions with Fc exhibited similar systemic pharmacological potency. The study demonstrates that modular incorporation of FC5 as the BBB-carrier arm in bispecific antibodies or antibody-drug conjugates offers an avenue to develop pharmacologically active biotherapeutics for CNS indications.


Subject(s)
Antibodies, Bispecific/metabolism , Biological Products/metabolism , Blood-Brain Barrier/metabolism , Animals , Antibodies, Bispecific/immunology , Biological Transport/physiology , Brain/metabolism , Humans , Immunoconjugates/metabolism , Male , Protein Engineering/methods , Rats, Wistar , Recombinant Fusion Proteins/metabolism
4.
J Struct Biol ; 185(2): 223-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23994046

ABSTRACT

A number of light and heavy chain canonical residue core redesigns were made in a therapeutic antibody (AQC2, anti-VLA1) Fab to explore the consequences to binding affinity and stability. These positions are all loop supporting, primarily CDR1 residues which do not directly contact the antigen. Structure based methods were used with and without consensus sequence information. 30 constructs were made, 24 expressed, and 70% of the designs using consensus sequence information retained binding affinity. Some success maintaining stability with more extreme redesigns suggests a surprising tolerance to mutation, though it often comes at the cost of loss of binding affinity and presumed loop conformation changes. In concordance with the expected need to present an ordered surface for binding, a relationship between decreased affinity and decreased stability was observed. Overpacking the core tends to destabilize the molecule and should be avoided.


Subject(s)
Complementarity Determining Regions/chemistry , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Light Chains/chemistry , Amino Acid Substitution , Animals , Antibody Affinity , Binding Sites , Complementarity Determining Regions/genetics , Humans , Hydrogen Bonding , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Integrin alpha1beta1/chemistry , Integrin alpha1beta1/immunology , Models, Molecular , Protein Binding , Protein Engineering , Protein Interaction Domains and Motifs , Protein Stability , Protein Unfolding , Rats , Thermodynamics
5.
Protein Sci ; 19(5): 954-66, 2010 May.
Article in English | MEDLINE | ID: mdl-20198683

ABSTRACT

Monoclonal antibodies (Mabs) are a favorite drug platform of the biopharmaceutical industry. Currently, over 20 Mabs have been approved and several hundred others are in clinical trials. The anti-LINGO-1 Mab Li33 was selected from a large panel of antibodies by Fab phage display technology based on its extraordinary biological activity in promoting oligodendrocyte differentiation and myelination in vitro and in animal models of remyelination. However, the Li33 Fab had poor solubility when converted into a full antibody in an immunoglobulin G1 framework. A detailed analysis of the biochemical and structural features of the antibody revealed several possible reasons for its propensity to aggregate. Here, we successfully applied three molecular approaches (isotype switching, targeted mutagenesis of complementarity determining region residues, and glycosylation site insertion mutagenesis) to address the solubility problem. Through these efforts we were able to improve the solubility of the Li33 Mab from 0.3 mg/mL to >50 mg/mL and reduce aggregation to an acceptable level. These strategies can be readily applied to other proteins with solubility issues.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Membrane Proteins/immunology , Mutagenesis, Site-Directed/methods , Nerve Tissue Proteins/immunology , Protein Engineering/methods , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Area Under Curve , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Immunoglobulin Class Switching , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Stability , Solubility , Temperature
6.
Protein Eng Des Sel ; 22(2): 93-101, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074157

ABSTRACT

A design approach was taken to investigate the feasibility of replacing single complementarity determining region (CDR) antibody loops. This approach may complement simpler mutation-based strategies for rational antibody design by expanding conformation space. Enormous crystal structure diversity is available, making CDR loops logical targets for structure-based design. A detailed analysis for the L1 loop shows that each loop length takes a distinct conformation, thereby allowing control on a length scale beyond that accessible to simple mutations. The L1 loop in the anti-VLA1 antibody was replaced with the L2 loop residues longer in an attempt to add an additional hydrogen bond and fill space on the antibody-antigen interface. The designs expressed well, but failed to improve affinity. In an effort to learn more, one design was crystallized and data were collected at 1.9 A resolution. The designed L1 loop takes the qualitatively desired conformation; confirming that loop replacement by design is feasible. The crystal structure also shows that the outermost loop (residues Leu51-Ser68) is domain swapped with another monomer. Tryptophan fluorescence measurements were used to monitor unfolding as a function of temperature and indicate that the loop involved in domain swapping does not unfold below 60 degrees C. The domain-swapping is not directly responsible for the affinity loss, but is likely a side-effect of the structural instability which may contribute to affinity loss. A second round of design was successful in eliminating the dimerization through mutation of a residue (Leu51Ser) at the joint of the domain-swapped loop.


Subject(s)
Antibodies/genetics , Complementarity Determining Regions/genetics , Immunoglobulin Fab Fragments/genetics , Animals , Antibodies/chemistry , Antibodies/metabolism , Antibody Affinity , Cloning, Molecular , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/metabolism , Computer Simulation , Crystallography, X-Ray , Escherichia coli/genetics , Feasibility Studies , Fluorescence , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Models, Molecular , Protein Conformation , Protein Engineering , Protein Folding , Protein Multimerization , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Temperature , Tryptophan/metabolism
7.
Biochemistry ; 45(33): 10117-28, 2006 Aug 22.
Article in English | MEDLINE | ID: mdl-16906770

ABSTRACT

LTbetaR is a member of the TNF receptor family of proteins. It binds to two different cell surface ligands, LIGHT, a homotypic trimer, and LTalpha1beta2, a heterotypic trimer. We have measured the affinities of the dimeric IgG fusion protein, LTbetaRIgG, and monomeric LTbetaR protein binding to both LIGHT and LTalpha1beta2 using surface plasmon resonance and found values of <0.1 and 38 nM for LIGHT and <0.1 and 48 nM for LTalpha1beta2, respectively. We also determined the stoichiometries of binding for both forms of the receptor LTbetaRIgG and LTbetaR binding to LIGHT. The data obtained from several biophysical methods are consistent with receptor polypeptide to trimeric ligand ratios of 2:1. The determined masses of the complexes using SEC-LS corresponded to a single LTbetaRIgG bound to a LIGHT trimer, or two LTbetaR bound per LIGHT. Sedimentation velocity of varied ratios of LTbetaR to a fixed concentration of LIGHT were analyzed by SEDANAL and were successfully fit with a model with two tight binding sites on LIGHT and one poor affinity site. Isothermal calorimetric titration of LIGHT with either LTbetaR or LTbetaRIgG also demonstrated stoichiometries of 1:2 and 1:1, respectively. The binding of LTbetaR to LIGHT was endothermic and, hence, entropy-driven. TNFR p55 (extracellular domain) complexed with the trimeric ligand, TNFbeta, exhibits a 3:1 receptor/ligand stoichiometry. This complex has been used as the prototypical model setting the receptor-ligand complexation paradigm for the entire TNF family. The LTbetaR/LIGHT binding stoichiometry of 2:1 demonstrated here does not fit the paradigm. This has numerous implications for cell biology including signaling requiring only dimerization of LTbetaR rather than trimerization as expected from the structural paradigm.


Subject(s)
Lymphotoxin-alpha/chemistry , Lymphotoxin-alpha/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism , Binding Sites , Calorimetry/methods , Cell Physiological Phenomena , Chromatography, Gel , Dimerization , Electrophoresis, Polyacrylamide Gel , Immunoglobulin G/metabolism , Ligands , Models, Molecular , Molecular Weight , Recombinant Fusion Proteins/metabolism , Surface Plasmon Resonance , Thermodynamics
8.
Protein Sci ; 15(5): 949-60, 2006 May.
Article in English | MEDLINE | ID: mdl-16597831

ABSTRACT

Improving the affinity of a high-affinity protein-protein interaction is a challenging problem that has practical applications in the development of therapeutic biomolecules. We used a combination of structure-based computational methods to optimize the binding affinity of an antibody fragment to the I-domain of the integrin VLA1. Despite the already high affinity of the antibody (Kd approximately 7 nM) and the moderate resolution (2.8 A) of the starting crystal structure, the affinity was increased by an order of magnitude primarily through a decrease in the dissociation rate. We determined the crystal structure of a high-affinity quadruple mutant complex at 2.2 A. The structure shows that the design makes the predicted contacts. Structural evidence and mutagenesis experiments that probe a hydrogen bond network illustrate the importance of satisfying hydrogen bonding requirements while seeking higher-affinity mutations. The large and diverse set of interface mutations allowed refinement of the mutant binding affinity prediction protocol and improvement of the single-mutant success rate. Our results indicate that structure-based computational design can be successfully applied to further improve the binding of high-affinity antibodies.


Subject(s)
Antibodies/therapeutic use , Antibody Affinity , Binding Sites, Antibody , Computer-Aided Design , Drug Design , Amino Acid Substitution , Antigen-Antibody Complex/chemistry , Crystallography, X-Ray , Immunoglobulins , Integrin alpha1beta1/immunology , Models, Molecular , Structure-Activity Relationship
9.
Biochemistry ; 45(7): 2006-13, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16475789

ABSTRACT

The oligomeric state of BAFF (B cell activing factor), a tumor necrosis factor (TNF) family cytokine that plays a critical role in B cell development and survival, has been the subject of recent debate. Myc-tagged BAFF starting at residue Gln136 was previously reported to crystallize as trimers at pH 4.5, whereas a histidine-tagged construct of BAFF, starting at residue Ala134, formed a virus-like cluster containing 60 monomers when crystallized at pH 9.0. The formation of the BAFF 60-mer was pH dependent, requiring pH >or= 7.0. More recently, 60-mer formation was suggested to be artificially induced by the histidine tag, and it was proposed that BAFF, like all other TNF family members, is trimeric. We report here that a construct of BAFF with no amino-terminal tag (Ala134-BAFF) can form a 60-mer in solution. Using size exclusion chromatography and static light scattering to monitor trimer to 60-mer ratios in BAFF preparations, we find that 60-mer formation is pH-dependent and requires histidine 218 within the DE loop of BAFF. Biacore measurements established that the affinity of Ala134-BAFF for the BAFF receptor BAFFR/BR3 is similar to that of myc-Gln136-BAFF, which is exclusively trimeric in solution. However, Ala134-BAFF is more efficacious than myc-Gln136-BAFF in inducing B cell proliferation in vitro. We additionally show that BAFF that is processed and secreted by 293T cells transfected with full-length BAFF, or by a histiocytic lymphoma cell line (U937) that expresses BAFF endogenously, forms a pH-dependent 60-mer in solution. Our results indicate that the formation of the 60-mer in solution by the BAFF extracellular domain is an intrinsic property of the protein, and therefore that this more active form of BAFF may be physiologically relevant.


Subject(s)
Membrane Proteins/physiology , Protein Structure, Quaternary , Tumor Necrosis Factor-alpha/physiology , Animals , B-Cell Activating Factor , Chromatography, Gel , Humans , Hydrogen-Ion Concentration , Light , Mice , Molecular Weight , Pichia/metabolism , Scattering, Radiation
10.
Science ; 310(5750): 1022-5, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16284179

ABSTRACT

We have identified a small-molecule inhibitor of tumor necrosis factor alpha (TNF-alpha) that promotes subunit disassembly of this trimeric cytokine family member. The compound inhibits TNF-alpha activity in biochemical and cell-based assays with median inhibitory concentrations of 22 and 4.6 micromolar, respectively. Formation of an intermediate complex between the compound and the intact trimer results in a 600-fold accelerated subunit dissociation rate that leads to trimer dissociation. A structure solved by x-ray crystallography reveals that a single compound molecule displaces a subunit of the trimer to form a complex with a dimer of TNF-alpha subunits.


Subject(s)
Indoles/chemistry , Indoles/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/chemistry , Biotinylation , Chemical Phenomena , Chemistry, Physical , Crystallography, X-Ray , Dimerization , Fluorescence , Hydrogen/chemistry , Hydrophobic and Hydrophilic Interactions , Indoles/chemical synthesis , Kinetics , Mass Spectrometry , Models, Chemical , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Conformation , Protein Subunits/chemistry , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL