Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(1): 101429, 2022 01.
Article in English | MEDLINE | ID: mdl-34801555

ABSTRACT

Noncovalent complexes of transforming growth factor-ß family growth/differentiation factors with their prodomains are classified as latent or active, depending on whether the complexes can bind their respective receptors. For the anti-Müllerian hormone (AMH), the hormone-prodomain complex is active, and the prodomain is displaced upon binding to its type II receptor, AMH receptor type-2 (AMHR2), on the cell surface. However, the mechanism by which this displacement occurs is unclear. Here, we used ELISA assays to measure the dependence of prodomain displacement on AMH concentration and analyzed results with respect to the behavior expected for reversible binding in combination with ligand-induced receptor dimerization. We found that, in solution, the prodomain has a high affinity for the growth factor (GF) (Kd = 0.4 pM). Binding of the AMH complex to a single AMHR2 molecule does not affect this Kd and does not induce prodomain displacement, indicating that the receptor binding site in the AMH complex is fully accessible to AMHR2. However, recruitment of a second AMHR2 molecule to bind the ligand bivalently leads to a 1000-fold increase in the Kd for the AMH complex, resulting in rapid release of the prodomain. Displacement occurs only if the AMHR2 is presented on a surface, indicating that prodomain displacement is caused by a conformational change in the GF induced by bivalent binding to AMHR2. In addition, we demonstrate that the bone morphogenetic protein 7 prodomain is displaced from the complex with its GF by a similar process, suggesting that this may represent a general mechanism for receptor-mediated prodomain displacement in this ligand family.


Subject(s)
Anti-Mullerian Hormone , Peptide Hormones , Anti-Mullerian Hormone/metabolism , Ligands , Peptide Hormones/metabolism , Protein Domains , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism
2.
MAbs ; 12(1): 1713648, 2020.
Article in English | MEDLINE | ID: mdl-31928294

ABSTRACT

LINGO-1 is a membrane protein of the central nervous system (CNS) that suppresses myelination of axons. Preclinical studies have revealed that blockade of LINGO-1 function leads to CNS repair in demyelinating animal models. The anti-LINGO-1 antibody Li81 (opicinumab), which blocks LINGO-1 function and shows robust remyelinating activity in animal models, is currently being investigated in a Phase 2 clinical trial as a potential treatment for individuals with relapsing forms of multiple sclerosis (AFFINITY: clinical trial.gov number NCT03222973). Li81 has the unusual feature that it contains two LINGO-1 binding sites: a classical site utilizing its complementarity-determining regions and a cryptic secondary site involving Li81 light chain framework residues that recruits a second LINGO-1 molecule only after engagement of the primary binding site. Concurrent binding at both sites leads to formation of a 2:2 complex of LINGO-1 with the Li81 antigen-binding fragment, and higher order complexes with intact Li81 antibody. To elucidate the role of the secondary binding site, we designed a series of Li81 variant constructs that eliminate it while retaining the classic site contacts. These Li81 mutants retained the high affinity binding to LINGO-1, but lost the antibody-induced oligodendrocyte progenitor cell (OPC) differentiation activity and myelination activity in OPC- dorsal root ganglion neuron cocultures seen with Li81. The mutations also attenuate antibody-induced internalization of LINGO-1 on cultured cortical neurons, OPCs, and cells over-expressing LINGO-1. Together these studies reveal that engagement at both LINGO-1 binding sites of Li81 is critical for robust functional activity of the antibody.


Subject(s)
Antibodies, Monoclonal/immunology , Binding Sites, Antibody/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/immunology , Humans
3.
Neurobiol Dis ; 124: 276-288, 2019 04.
Article in English | MEDLINE | ID: mdl-30381260

ABSTRACT

Aggregation of α-synuclein (α-syn) is neuropathologically and genetically linked to Parkinson's disease (PD). Since stereotypic cell-to-cell spreading of α-syn pathology is believed to contribute to disease progression, immunotherapy with antibodies directed against α-syn is considered a promising therapeutic approach for slowing disease progression. Here we report the identification, binding characteristics, and efficacy in PD mouse models of the human-derived α-syn antibody BIIB054, which is currently under investigation in a Phase 2 clinical trial for PD. BIIB054 was generated by screening human memory B-cell libraries from healthy elderly individuals. Epitope mapping studies conducted using peptide scanning, X-ray crystallography, and mutagenesis show that BIIB054 binds to α-syn residues 1-10. BIIB054 is highly selective for aggregated forms of α-syn with at least an 800-fold higher apparent affinity for fibrillar versus monomeric recombinant α-syn and a strong preference for human PD brain tissue. BIIB054 discriminates between monomers and oligomeric/fibrillar forms of α-syn based on high avidity for aggregates, driven by weak monovalent affinity and fast binding kinetics. In efficacy studies in three different mouse models with intracerebrally inoculated preformed α-syn fibrils, BIIB054 treatment attenuated the spreading of α-syn pathology, rescued motor impairments, and reduced the loss of dopamine transporter density in dopaminergic terminals in striatum. The preclinical data reported here provide a compelling rationale for clinical development of BIIB054 for the treatment and prevention of PD.


Subject(s)
Antibodies, Monoclonal/pharmacology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , alpha-Synuclein/antagonists & inhibitors , Animals , Humans , Mice , Phenotype , Protein Aggregates
4.
Sci Rep ; 8(1): 6412, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686315

ABSTRACT

Aducanumab, a human-derived antibody targeting amyloid-ß (Aß), is in Phase 3 clinical trials for the treatment of Alzheimer's disease. Biochemical and structural analyses show that aducanumab binds a linear epitope formed by amino acids 3-7 of the Aß peptide. Aducanumab discriminates between monomers and oligomeric or fibrillar aggregates based on weak monovalent affinity, fast binding kinetics and strong avidity for epitope-rich aggregates. Direct comparative studies with analogs of gantenerumab, bapineuzumab and solanezumab demonstrate clear differentiation in the binding properties of these antibodies. The crystal structure of the Fab fragment of aducanumab bound to its epitope peptide reveals that aducanumab binds to the N terminus of Aß in an extended conformation, distinct from those seen in structures with other antibodies that target this immunodominant epitope. Aducanumab recognizes a compact epitope that sits in a shallow pocket on the antibody surface. In silico analyses suggest that aducanumab interacts weakly with the Aß monomer and may accommodate a variety of peptide conformations, further supporting its selectivity for Aß aggregates. Our studies provide a structural rationale for the low affinity of aducanumab for non-pathogenic monomers and its greater selectivity for aggregated forms than is seen for other Aß-targeting antibodies.


Subject(s)
Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Binding Sites, Antibody , Enzyme-Linked Immunosorbent Assay , Humans , Immunotherapy , Kinetics , Molecular Docking Simulation , Protein Conformation , Surface Plasmon Resonance
5.
Anal Chem ; 89(7): 4021-4030, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28245108

ABSTRACT

Meteorin and Cometin (Meteorin-like) are secreted proteins belonging to a newly discovered growth factor family. Both proteins play important roles in neural development and may have potential as therapeutic targets or agents. Meteorin and Cometin are homologues and contain ten evolutionarily conserved Cys residues across a wide variety of species. However, the status of the Cys residues has remained unknown. Here, we have successfully determined the disulfide structure for murine Meteorin by LC-MS analysis of fragments generated by trypsin plus endoprotease-Asp-N. For proteolytic fragments linked by more than one disulfide bond, we used electron transfer dissociation (ETD) to partially dissociate disulfide bonds followed by high-energy collisional dissociation (HCD) to determine disulfide linkages. Our analysis revealed that the ten Cys residues in murine Meteorin form five disulfide bonds with Cys7 (C1) linked to Cys28 (C2), Cys59 (C3) to Cys95 (C4), Cys148 (C5) to Cys219 (C8), Cys151 (C6) to Cys243 (C9), and Cys161 (C7) to Cys266 (C10). Since the ten Cys residues are highly conserved in Meteorin and Cometin, it is likely that the disulfide linkages are also conserved. This disulfide structure information should facilitate structure-function relationship studies on this new class of neurotrophic factors and also assist in evaluation of their therapeutic potentials.


Subject(s)
Disulfides/analysis , Nerve Growth Factors/chemistry , Peptide Fragments/chemistry , Proteolysis , Animals , Chromatography, Liquid , Electron Transport , Energy Transfer , Mice , Molecular Structure , Tandem Mass Spectrometry
6.
J Clin Endocrinol Metab ; 101(12): 4618-4627, 2016 12.
Article in English | MEDLINE | ID: mdl-27623067

ABSTRACT

CONTEXT: Anti-Müllerian hormone (AMH) is an important clinical marker for diagnosing and assessing the reproductive status and/or disorders in men and women. Most studies have not distinguished between levels of inactive AMH precursor and the cleaved noncovalent complex that binds the AMH type II receptor (AMHRII) and initiates signaling. OBJECTIVE: The objective of the study was to measure the levels of AMH cleavage and bioactivity in human body fluids. DESIGN, SETTING, AND PATIENTS: AMH cleavage levels and bioactivity were measured in the serum of six boys and in the follicular fluid and serum of nine control women and 13 women with the polycystic ovary syndrome (PCOS). MAIN OUTCOME MEASURES: AMH cleavage levels were measured by capturing AMH with an anti-AMH antibody, followed by Western blotting. The bioactivity of cleaved AMH was assessed with an ELISA that measures the levels of AMH capable of binding AMHRII. RESULTS: PCOS women have an elevated level of AMH cleavage in their follicular fluid (24% vs 8% in control women), and most of the cleaved AMH can bind AMHRII. Higher levels of cleavage are observed in female (60%) and male (79%) serum, but very little of the cleaved AMH can bind AMHRII. CONCLUSIONS: These results support an autocrine role for AMH in the pathophysiology of PCOS in the follicle. In addition, they indicate that AMH undergoes interactions or structural changes after cleavage that prevent receptor binding, meaning, unexpectedly, that the level of cleaved AMH in biological fluids does not always reflect the level of bioactive AMH.


Subject(s)
Anti-Mullerian Hormone/metabolism , Follicular Fluid/metabolism , Polycystic Ovary Syndrome/metabolism , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Adult , Anti-Mullerian Hormone/blood , Child , Female , Humans , Male , Polycystic Ovary Syndrome/blood , Protein Binding
7.
J Cell Sci ; 128(7): 1352-64, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25663701

ABSTRACT

The levels and intracellular localization of wild-type transforming growth factor ß superfamily (TGFß-SF) receptors are tightly regulated by endocytic trafficking, shedding and degradation. In contrast, a main regulatory mechanism of mutation-bearing receptors involves their intracellular retention. Anti-Müllerian hormone receptor II (AMHRII, also known as AMHR2) is the type-II receptor for anti-Müllerian hormone (AMH), a TGFß-SF ligand that mediates Müllerian duct regression in males. Here, we studied AMHRII processing and identified novel mechanisms of its constitutive negative regulation. Immunoblot analysis revealed that a significant portion of AMHRII was missing most of its extracellular domain (ECD) and, although glycosylated, was unfolded and retained in the endoplasmic reticulum. Exogenous expression of AMHRII, but not of type-II TGF-ß receptor (TßRII, also known as TGFR2), resulted in its disulfide-bond-mediated homo-oligomerization and intracellular retention, and in a decrease in its AMH-binding capacity. At the plasma membrane, AMHRII differed from TßRII, forming high levels of non-covalent homomeric complexes, which exhibited a clustered distribution and restricted lateral mobility. This study identifies novel mechanisms of negative regulation of a type-II TGFß-SF receptor through cleavage, intracellular retention and/or promiscuous disulfide-bond mediated homo-oligomerization.


Subject(s)
Protein Processing, Post-Translational , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Anti-Mullerian Hormone/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Male , Mice , Protein Binding , Protein Folding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Receptor, Transforming Growth Factor-beta Type II , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
8.
J Pharmacol Exp Ther ; 350(1): 110-23, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24756303

ABSTRACT

Multiple sclerosis (MS) is an autoimmune-inflammatory disease of the central nervous system (CNS) with prominent demyelination and axonal injury. While most MS therapies target the immunologic response, there is a large unmet need for treatments that can promote CNS repair. LINGO-1 (leucine-rich repeat and Ig-containing Nogo receptor interacting protein-1) is a membrane protein selectively expressed in the CNS that suppresses myelination, preventing the repair of damaged axons. We are investigating LINGO-1 antagonist antibodies that lead to remyelination as a new paradigm for treatment of individuals with MS. The anti-LINGO-1 Li81 antibody,BIIB033, is currently in clinical trials and is the first MS treatment targeting CNS repair. Here, to elucidate the mechanism of action of the antibody, we solved the crystal structure of the LINGO-1-Li81 Fab complex and used biochemical and functional studies to investigate structure-function relationships. Li81 binds to the convex surface of the leucine-rich repeat domain of LINGO-1 within repeats 4-8. Fab binding blocks contact points used in the oligomerization of LINGO-1 and produces a stable complex containing two copies each of LINGO-1 and Fab that results from a rearrangement of contacts stabilizing the quaternary structure of LINGO-1. The formation of the LINGO-1-Li81 Fab complex masks functional epitopes within the Ig domain of LINGO-1 that are important for its biologic activity in oligodendrocyte differentiation. These studies provide new insights into the structure and biology of LINGO-1 and how Li81 monoclonal antibody can block its function.


Subject(s)
Antibodies, Monoclonal/metabolism , Immunoglobulin Fab Fragments/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Differentiation/immunology , Female , Humans , Molecular Structure , Multiple Sclerosis/drug therapy , Oligodendroglia/immunology , Protein Binding , Protein Structure, Quaternary , Rats
9.
Methods ; 65(1): 68-76, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23816785

ABSTRACT

Antibodies are key components of the adaptive immune system and are well-established protein therapeutic agents. Typically high-affinity antibodies are obtained by immunization of rodent species that need to be humanized to reduce their immunogenicity. The complementarity-determining regions (CDRs) contain the residues in a defined loop structure that confer antigen binding, which must be retained in the humanized antibody. To design a humanized antibody, we graft the mature murine CDRs onto a germline human acceptor framework. Structural defects due to mismatches at the graft interface can be fixed by mutating some framework residues to murine, or by mutating some residues on the CDRs' backside to human or to a de novo designed sequence. The first approach, framework redesign, can yield an antibody with binding better than the CDR graft and one equivalent to the mature murine, and reduced immunogenicity. The second approach, CDR redesign, is presented here as a new approach, yielding an antibody with binding better than the CDR graft, and immunogenicity potentially less than that from framework redesign. Application of both approaches to the humanization of anti-α4 integrin antibody HP1/2 is presented and the concept of the hybrid humanization approach that retains "difficult to match" murine framework amino acids and uses de novo CDR design to minimize murine amino acid content and reduce cell-mediated cytotoxicity liabilities is discussed.


Subject(s)
Antibodies, Monoclonal, Humanized/biosynthesis , Complementarity Determining Regions/biosynthesis , Immunoglobulin Fab Fragments/biosynthesis , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibody Affinity , Binding Sites , Cloning, Molecular , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Hybridomas , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Jurkat Cells , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed
10.
CNS Drugs ; 27(7): 493-503, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23681979

ABSTRACT

LINGO-1 is a leucine-rich repeat and Ig domain-containing, Nogo receptor interacting protein, selectively expressed in the CNS on both oligodendrocytes and neurons. Its expression is developmentally regulated, and is upregulated in CNS diseases and injury. In animal models, LINGO-1 expression is upregulated in rat spinal cord injury, experimental autoimmune encephalomyelitis, 6-hydroxydopamine neurotoxic lesions and glaucoma models. In humans, LINGO-1 expression is increased in oligodendrocyte progenitor cells from demyelinated white matter of multiple sclerosis post-mortem samples, and in dopaminergic neurons from Parkinson's disease brains. LINGO-1 negatively regulates oligodendrocyte differentiation and myelination, neuronal survival and axonal regeneration by activating ras homolog gene family member A (RhoA) and inhibiting protein kinase B (Akt) phosphorylation signalling pathways. Across diverse animal CNS disease models, targeted LINGO-1 inhibition promotes neuron and oligodendrocyte survival, axon regeneration, oligodendrocyte differentiation, remyelination and functional recovery. The targeted inhibition of LINGO-1 function presents a novel therapeutic approach for the treatment of CNS diseases.


Subject(s)
Central Nervous System Diseases/drug therapy , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/drug effects , Oligodendroglia/drug effects , Aging/metabolism , Aging/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Cell Survival/drug effects , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Clinical Trials as Topic , Humans , Membrane Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Signal Transduction
11.
J Biol Chem ; 287(39): 32897-912, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22847004

ABSTRACT

We have applied hydrogen-deuterium exchange mass spectrometry, in conjunction with differential scanning calorimetry and protein stability analysis, to examine solution dynamics of the integrin α1 I domain induced by the binding of divalent cations, full-length type IV collagen, or a function-blocking monoclonal antibody. These studies revealed features of integrin activation and α1I-ligand complexes that were not detected by static crystallographic data. Mg(2+) and Mn(2+) stabilized α1I but differed in their effects on exchange rates in the αC helix. Ca(2+) impacted α1I conformational dynamics without altering its gross thermal stability. Interaction with collagen affected the exchange rates in just one of three metal ion-dependent adhesion site (MIDAS) loops, suggesting that MIDAS loop 2 plays a primary role in mediating ligand binding. Collagen also induced changes consistent with increased unfolding in both the αC and allosteric C-terminal helices of α1I. The antibody AQC2, which binds to α1I in a ligand-mimetic manner, also reduced exchange in MIDAS loop 2 and increased exchange in αC, but it did not impact the C-terminal region. This is the first study to directly demonstrate the conformational changes induced upon binding of an integrin I domain to a full-length collagen ligand, and it demonstrates the utility of the deuterium exchange mass spectrometry method to study the solution dynamics of integrin/ligand and integrin/metal ion interactions. Based on the ligand and metal ion binding data, we propose a model for collagen-binding integrin activation that explains the differing abilities of Mg(2+), Mn(2+), and Ca(2+) to activate I domain-containing integrins.


Subject(s)
Collagen Type IV/metabolism , Integrin alpha1/metabolism , Magnesium/metabolism , Manganese/metabolism , Animals , Collagen Type IV/chemistry , Humans , Integrin alpha1/chemistry , Integrin alpha1/genetics , Magnesium/chemistry , Manganese/chemistry , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Rats
12.
J Pharmacol Exp Ther ; 339(2): 519-29, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21807883

ABSTRACT

LINGO-1 (leucine-rich repeat and Ig domain containing NOGO receptor interacting protein-1) is a negative regulator of myelination and repair of damaged axons in the central nervous system (CNS). Blocking LINGO-1 function leads to robust remyelination. The anti-LINGO-1 Li81 antibody is currently being evaluated in clinical trials for multiple sclerosis (MS) and is the first MS therapy that directly targets myelin repair. LINGO-1 is selectively expressed in brain and spinal cord but not in peripheral tissues. Perhaps the greatest concern for Li81 therapy is the limited access of the drug to the CNS. Here, we measured Li81 concentrations in brain, spinal cord, and cerebral spinal fluid in rats after systemic administration and correlated them with dose-efficacy responses in rat lysolecithin and experimental autoimmune encephalomyelitis spinal cord models of remyelination. Remyelination was dose-dependent, and levels of Li81 in spinal cord that promoted myelination correlated well with affinity measurements for the binding of Li81 to LINGO-1. Observed Li81 concentrations in the CNS of 0.1 to 0.4% of blood levels are consistent with values reported for other antibodies. To understand the features of the antibody that affect CNS penetration, we also evaluated the pharmacokinetics of Li81 Fab2, Fab, and poly(ethylene glycol)-modified Fab. The reagents all showed similar CNS exposure despite large differences in their sizes, serum half-lives, and volumes of distribution, and area under the curve (AUC) measurements in the CNS directly correlated with AUC measurements in serum. These studies demonstrate that exposure levels achieved by passive diffusion of the Li81 monoclonal antibody into the CNS are sufficient and lead to robust remyelination.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/immunology , Spinal Cord/drug effects , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Cerebrospinal Fluid/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Lysophosphatidylcholines , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Regeneration , Spinal Cord/metabolism , Spinal Cord/pathology
13.
Mol Cell Neurosci ; 48(1): 72-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21740973

ABSTRACT

The sphingosine 1-phosphate (S1P) receptor modulators have emerged as a new therapeutic opportunity paradigm for the treatment of immune-mediated demyelinating diseases such as multiple sclerosis (MS). The S1P analog fingolimod (FTY720) has been shown to alleviate disease burden in immune-mediated animal models of MS, and has been approved for treatment in clinical trials in patients with MS in the United States. While the immunological effects of FTY720 are well established, there is controversy in the literature regarding the contribution of FTY720 on myelin repair. Here, we directly assessed the impact of FTY720 on myelin repair in cuprizone and lysolecithin (LPC) demyelination models that have a minimal immunological component. FTY720 failed to promote remyelination in either animal model. These studies suggest that while FTY720 may be effective at modulating the immunological attack in MS, it may benefit from an add-on therapy to enhance the myelin repair required for long-term functional restoration in MS.


Subject(s)
Immunosuppressive Agents/pharmacology , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Nerve Regeneration/drug effects , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Chelating Agents/pharmacology , Cuprizone/pharmacology , Demyelinating Diseases/drug therapy , Disease Models, Animal , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/therapeutic use , Mice , Mice, Inbred C57BL , Propylene Glycols/therapeutic use , Rats , Rats, Sprague-Dawley , Sphingosine/pharmacology , Sphingosine/therapeutic use
14.
Nat Med ; 17(7): 816-21, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725297

ABSTRACT

Survival and differentiation of oligodendrocytes are important for the myelination of central nervous system (CNS) axons during development and crucial for myelin repair in CNS demyelinating diseases such as multiple sclerosis. Here we show that death receptor 6 (DR6) is a negative regulator of oligodendrocyte maturation. DR6 is expressed strongly in immature oligodendrocytes and weakly in mature myelin basic protein (MBP)-positive oligodendrocytes. Overexpression of DR6 in oligodendrocytes leads to caspase 3 (casp3) activation and cell death. Attenuation of DR6 function leads to enhanced oligodendrocyte maturation, myelination and downregulation of casp3. Treatment with a DR6 antagonist antibody promotes remyelination in both lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis (EAE) models. Consistent with the DR6 antagoinst antibody studies, DR6-null mice show enhanced remyelination in both demyelination models. These studies reveal a pivotal role for DR6 signaling in immature oligodendrocyte maturation and myelination that may provide new therapeutic avenues for the treatment of demyelination disorders such as multiple sclerosis.


Subject(s)
Myelin Sheath/metabolism , Oligodendroglia/metabolism , Receptors, Tumor Necrosis Factor/physiology , Animals , Blotting, Western , Caspase 3/metabolism , Caspase 3/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Survival/physiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Enzyme Activation , Gene Expression Regulation , Mice , Multiple Sclerosis/metabolism , Multiple Sclerosis/therapy , Myelin Sheath/physiology , Oligodendroglia/physiology , Rats , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
J Pharm Biomed Anal ; 55(1): 168-75, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21300512

ABSTRACT

Natalizumab is a humanized IgG4 monoclonal antibody which binds human α4 integrin and is approved for treatment of multiple sclerosis and Crohn's disease. Assessment of the in vivo disposition of natalizumab presents a unique assay development challenge due to the ability of human IgG4 antibodies to undergo half-antibody exchange in vivo. Such exchange generates IgG4 molecules of mixed specificity comprising a natalizumab heavy-light chain pair coupled to an IgG4 heavy-light chain pair of unknown specificity. Since exchanged and non-exchanged species cannot be quantified independently using a single enzyme linked immunosorbent assay (ELISA), a novel quantitation strategy was developed employing two ELISAs: one measuring total natalizumab including both intact and exchanged molecules, and the second measuring only intact natalizumab. The presence and amount of exchanged natalizumab in serum is calculated by the difference in values obtained in the two assays. To evaluate assay performance, a control reagent was created from natalizumab and an irrelevant humanized monoclonal IgG4 antibody. Subsequent validation demonstrated that both assays are specific, accurate, and precise within the working ranges of the assays (1.5-10µg/mL for total and 0.5-12µg/mL for intact natalizumab assays). The mean accuracy, intra- and inter-assay precision for both assays were 82-113%, ≤9% and ≤20%, respectively. Additionally, the limits of detection of intact and exchanged natalizumab were established using statistical methods. The utility of the two-assay strategy was confirmed by analyzing samples from a pharmacokinetic study in rats using different variants of natalizumab administered along with another human IgG4 antibody as an exchange partner.


Subject(s)
Antibodies, Monoclonal/metabolism , Immunoassay/methods , Immunoglobulin G/metabolism , Immunoglobulin Variable Region/metabolism , Integrin alpha4/metabolism , Algorithms , Animals , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Crohn Disease/drug therapy , Humans , Immunoglobulin G/blood , Immunoglobulin Variable Region/immunology , Limit of Detection , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Mutant Proteins/metabolism , Mutant Proteins/pharmacokinetics , Natalizumab , Protein Refolding , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Technology, Pharmaceutical
16.
Bioconjug Chem ; 22(2): 200-10, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21254764

ABSTRACT

The use of LINGO-1 antagonists to promote repair of damaged myelin is an emerging therapeutic opportunity for treatment of CNS diseases caused by demyelination such as multiple sclerosis. The Li33 anti-LINGO-1 antibody is a potent inducer of myelination in vitro and in vivo, but aggregation issues prevented the engineering of an optimal development candidate. PEGylated Li33 Fab' is one of several versions of the Li33 antibody that is being investigated in an attempt to identify the most favorable anti-LINGO-1 antibody design. For targeted PEGylation, a Li33 Fab' construct was engineered with a single unpaired cysteine in the heavy-chain hinge sequence. The Fab' was expressed in CHO cells, purified, and PEGylated with 20 kDa methoxy-poly(ethylene glycol) maleimide using a reaction strategy optimized to improve the yield of the PEG-Fab'. Biochemical analysis of the Li33 PEG-Fab' verified the selectivity of the PEGylation reaction. The in vitro and in vivo attributes of the PEG-Fab' were benchmarked against a Li33 full antibody. Both the Li33 PEG-Fab' and intact antibody bound LINGO-1 with nanomolar affinity, promoted myelination in an in vitro signaling assay, and promoted the repair of damaged myelin in the rat lysolecithin model. These studies extend our understanding of the biological activity of the Li33 mAb and validate the use of an anti-LINGO-1 PEG-Fab' for treatment of CNS diseases caused by demyelination.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin Fab Fragments/chemistry , Polyethylene Glycols/chemistry , Animals , Antibodies, Monoclonal/immunology , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/isolation & purification , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/immunology , Models, Animal , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Rats
17.
Biotechnol Appl Biochem ; 57(1): 31-45, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20815818

ABSTRACT

NgRI (Nogo-66 receptor) is part of a signalling complex that inhibits axon regeneration in the central nervous system. Truncated soluble versions of NgRI have been used successfully to promote axon regeneration in animal models of spinal-cord injury, raising interest in this protein as a potential therapeutic target. The LRR (leucine-rich repeat) regions in NgRI are flanked by N- and C-terminal disulfide-containing 'cap' domains (LRRNT and LRRCT respectively). In the present work we show that, although functionally active, the NgRI(310)-Fc fusion protein contains mislinked and heterogeneous disulfide patterns in the LRRCT domain, and we report the generation of a series of variant molecules specifically designed to prevent this heterogeneity. Using these variants we explored the effects of modifying the NgRI truncation site or the spacing between the NgRI and Fc domains, or replacing cysteines within the NgRI or IgG hinge regions. One variant, which incorporates replacements of Cys²66 and Cys³°9 with alanine residues, completely eliminated disulfide scrambling while maintaining functional in vitro and in vivo efficacy. This modified NgRI-Fc molecule represents a significantly improved candidate for further pharmaceutical development, and may serve as a useful model for the optimization of other IgG fusion proteins made from LRR proteins.


Subject(s)
Disulfides/metabolism , Myelin Proteins/chemistry , Protein Engineering/methods , Receptors, Cell Surface/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Behavior, Animal/drug effects , Crystallography, X-Ray , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , Humans , Male , Molecular Sequence Annotation , Molecular Sequence Data , Myelin Proteins/genetics , Nogo Receptor 1 , Protein Stability , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Spinal Cord Injuries/drug therapy , Spinal Nerve Roots/injuries
18.
Mol Endocrinol ; 24(11): 2193-206, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20861221

ABSTRACT

TGF-ß family ligands are translated as prepropeptide precursors and are processed into mature C-terminal dimers that signal by assembling a serine/threonine kinase receptor complex containing type I and II components. Many TGF-ß ligands are secreted in a latent form that cannot bind their receptor, due to the pro-region remaining associated with the mature ligand in a noncovalent complex after proteolytic cleavage. Here we show that anti-Müllerian hormone (AMH), a TGF-ß family ligand involved in reproductive development, must be cleaved to bind its type II receptor (AMHRII), but dissociation of the pro-region from the mature C-terminal dimer is not required for this initial interaction. We provide direct evidence for this interaction by showing that the noncovalent complex binds to a soluble form of AMHRII in an ELISA format and to AMHRII immobilized on Sepharose. Binding of the noncovalent complex to Sepharose-coupled AMHRII induces dissociation of the pro-region from the mature C-terminal dimer, whereas no dissociation occurs after binding to immobilized AMH antibodies. The pro-region cannot be detected after binding of the AMH noncovalent complex to AMHRII expressed on COS cells, indicating that pro-region dissociation may occur as a natural consequence of receptor engagement on cells. Moreover, the mature C-terminal dimer is more active than the noncovalent complex in stimulating Sma- and Mad-related protein activation, suggesting that pro-region dissociation contributes to the assembly of the active receptor complex. AMH thus exemplifies a new mechanism for receptor engagement in which interaction with the type II receptor promotes pro-region dissociation to generate mature ligand.


Subject(s)
Anti-Mullerian Hormone/metabolism , Protein Processing, Post-Translational , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Anti-Mullerian Hormone/chemistry , COS Cells , Chlorocebus aethiops , Humans , Models, Biological , Phosphorylation , Protein Multimerization , Receptors, Fc/metabolism , Solubility
19.
J Chromatogr A ; 1217(32): 5225-32, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20598700

ABSTRACT

Protein trisulfide linkages are generated by the post-translational insertion of a sulfur atom into a disulfide bond. Molecular heterogeneity was detected in a recombinant IgG(1) monoclonal antibody (mAb) and attributed to the presence of a protein trisulfide moiety. The predominant site of trisulfide modification was the bond between the heavy and light chains. The trisulfide was eliminated during purification of the IgG(1) mAb via a cysteine wash step incorporated into Protein A affinity column chromatography. Analysis of the cysteine-treated mAb by electrophoresis and peptide mapping indicated that the trisulfide linkages were efficiently converted to intact disulfide bonds (13% trisulfide decreased consistently to 1% or less) without disulfide scrambling or an increase in free sulfhydryls. The on-column trisulfide conversion caused no change in protein folding detectable by hydrogen/deuterium exchange or differential scanning calorimetry. Consistent with this, binding of the mAb to its antigen in vitro was insensitive to the presence of the trisulfide modification and to its removal by the on-column cysteine treatment. Similar, high efficiency trisulfide conversion was achieved for a second IgG(1) mAb using the column wash strategy (at least 7% trisulfide decreased to 1% or less). Therefore, trisulfide/disulfide heterogeneity can be eliminated from IgG(1) molecules via a convenient and inexpensive procedure compatible with routine Protein A affinity capture.


Subject(s)
Antibodies, Monoclonal/chemistry , Chromatography, Affinity/methods , Immunoglobulin G/chemistry , Staphylococcal Protein A/metabolism , Sulfides/chemistry , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , CHO Cells , Calorimetry, Differential Scanning , Cricetinae , Cricetulus , Cysteine/chemistry , Cysteine/metabolism , Deuterium Exchange Measurement , Electrophoresis, Polyacrylamide Gel , Humans , Immunoglobulin G/isolation & purification , Immunoglobulin G/metabolism , Mass Spectrometry , Oxidation-Reduction , Peptide Mapping/methods , Staphylococcal Protein A/chemistry , Sulfides/metabolism , Time Factors
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...