Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Elife ; 72018 07 25.
Article in English | MEDLINE | ID: mdl-30039798

ABSTRACT

The early-life intestinal microbiota plays a key role in shaping host immune system development. We found that a single early-life antibiotic course (1PAT) accelerated type 1 diabetes (T1D) development in male NOD mice. The single course had deep and persistent effects on the intestinal microbiome, leading to altered cecal, hepatic, and serum metabolites. The exposure elicited sex-specific effects on chromatin states in the ileum and liver and perturbed ileal gene expression, altering normal maturational patterns. The global signature changes included specific genes controlling both innate and adaptive immunity. Microbiome analysis revealed four taxa each that potentially protect against or accelerate T1D onset, that were linked in a network model to specific differences in ileal gene expression. This simplified animal model reveals multiple potential pathways to understand pathogenesis by which early-life gut microbiome perturbations alter a global suite of intestinal responses, contributing to the accelerated and enhanced T1D development.


Subject(s)
Anti-Bacterial Agents/adverse effects , Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome/immunology , Immunity, Innate/drug effects , Adaptive Immunity/drug effects , Animals , Anti-Bacterial Agents/immunology , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/pathology , Female , Gastrointestinal Microbiome/drug effects , Ileum/immunology , Ileum/microbiology , Immunity, Innate/immunology , Intestines/microbiology , Mice , Mice, Inbred NOD , Microbiota/drug effects , Microbiota/immunology
2.
PLoS Pathog ; 14(3): e1006935, 2018 03.
Article in English | MEDLINE | ID: mdl-29509814

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract infections in infants and the elderly, and yet there remains no effective treatment or vaccine. The surface of the virion is decorated with the fusion glycoprotein (RSV F) and the attachment glycoprotein (RSV G), which binds to CX3CR1 on human airway epithelial cells to mediate viral attachment and subsequent infection. RSV G is a major target of the humoral immune response, and antibodies that target the central conserved region of G have been shown to neutralize both subtypes of RSV and to protect against severe RSV disease in animal models. However, the molecular underpinnings for antibody recognition of this region have remained unknown. Therefore, we isolated two human antibodies directed against the central conserved region of RSV G and demonstrated that they neutralize RSV infection of human bronchial epithelial cell cultures in the absence of complement. Moreover, the antibodies protected cotton rats from severe RSV disease. Both antibodies bound with high affinity to a secreted form of RSV G as well as to a peptide corresponding to the unglycosylated central conserved region. High-resolution crystal structures of each antibody in complex with the G peptide revealed two distinct conformational epitopes that require proper folding of the cystine noose located in the C-terminal part of the central conserved region. Comparison of these structures with the structure of fractalkine (CX3CL1) alone or in complex with a viral homolog of CX3CR1 (US28) suggests that RSV G would bind to CX3CR1 in a mode that is distinct from that of fractalkine. Collectively, these results build on recent studies demonstrating the importance of RSV G in antibody-mediated protection from severe RSV disease, and the structural information presented here should guide the development of new vaccines and antibody-based therapies for RSV.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/chemistry , Animals , Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Bronchi/drug effects , Bronchi/immunology , Bronchi/metabolism , Cells, Cultured , Chemokine CX3CL1/metabolism , Crystallography, X-Ray , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epitopes/chemistry , Epitopes/immunology , Humans , Male , Protein Conformation , Rats , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/pharmacology , Respiratory System/drug effects , Respiratory System/immunology , Respiratory System/metabolism , Sigmodontinae , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism
3.
J Infect Dis ; 211(2): 216-25, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-24864121

ABSTRACT

BACKGROUND: Human metapneumovirus (HMPV) is a leading cause of acute respiratory tract infection, with significant morbidity and mortality. No licensed vaccines or therapeutic agents exist. Monoclonal antibodies (mAbs) are effective at preventing other infectious diseases and could be used against HMPV in high-risk hosts. METHODS: In vitro assays were performed to assess the neutralizing activity and affinity kinetics of human mAb 54G10. A new mouse model was developed to assess prophylactic and therapeutic efficacy in vivo. The epitope of 54G10 was identified by generating mAb-resistant mutants (MARMs). RESULTS: At low concentrations, 54G10 neutralized all 4 subgroups of HMPV in vitro and had subnanomolar affinity for the fusion protein. DBA/2 mice were permissive for all 4 HMPV subgroups, and 54G10 was effective both prophylactically and therapeutically against HMPV in vivo. Sequencing of HMPV MARMs identified the 54G10 epitope, which was similar to an antigenic site on respiratory syncytial virus (RSV). 54G10 also exhibited in vitro neutralizing activity and in vivo protective and therapeutic efficacy against RSV. CONCLUSIONS: Human mAb 54G10 has broad neutralizing activity against HMPV and could have prophylactic and therapeutic utility clinically. The conserved epitope could represent a structural vaccine target for HMPV and RSV.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Metapneumovirus/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Antibodies, Monoclonal, Humanized/isolation & purification , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Chemoprevention/methods , Disease Models, Animal , Female , Humans , Immunotherapy/methods , Mice, Inbred BALB C , Mice, Inbred DBA , Neutralization Tests , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/therapy , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/therapy , Treatment Outcome
4.
J Virol ; 87(17): 9501-10, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23785217

ABSTRACT

In most forms of prion disease, infectivity is present primarily in the central nervous system or immune system organs such as spleen and lymph node. However, a transgenic mouse model of prion disease has demonstrated that prion infectivity can also be present as amyloid deposits in heart tissue. Deposition of infectious prions as amyloid in human heart tissue would be a significant public health concern. Although abnormal disease-associated prion protein (PrP(Sc)) has not been detected in heart tissue from several amyloid heart disease patients, it has been observed in the heart tissue of a patient with sporadic Creutzfeldt-Jakob Disease (sCJD), the most common form of human prion disease. In order to determine whether prion infectivity can be found in heart tissue, we have inoculated formaldehyde fixed brain and heart tissue from two sCJD patients, as well as prion protein positive fixed heart tissue from two amyloid heart disease patients, into transgenic mice overexpressing the human prion protein. Although the sCJD brain samples led to clinical or subclinical prion infection and deposition of PrP(Sc) in the brain, none of the inoculated heart samples resulted in disease or the accumulation of PrP(Sc). Thus, our results suggest that prion infectivity is not likely present in cardiac tissue from sCJD or amyloid heart disease patients.


Subject(s)
Amyloidosis/metabolism , Amyloidosis/pathology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Myocardium/metabolism , Myocardium/pathology , PrPSc Proteins/metabolism , PrPSc Proteins/pathogenicity , Animals , Brain/metabolism , Brain/pathology , Creutzfeldt-Jakob Syndrome/transmission , Cricetinae , Disease Models, Animal , Humans , Immunohistochemistry , Mice , Mice, Transgenic
5.
Ann Neurol ; 65(6): 639-49, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19557869

ABSTRACT

OBJECTIVE: Intrathecal IgG synthesis, persistence of bands of oligoclonal IgG, and memory B-cell clonal expansion are well-characterized features of the humoral response in multiple sclerosis (MS). Nevertheless, the target antigen of this response remains enigmatic. METHODS: We produced 53 different human IgG1 monoclonal recombinant antibodies (rAbs) by coexpressing paired heavy- and light-chain variable region sequences of 51 plasma cell clones and 2 B-lymphocyte clones from MS cerebrospinal fluid in human tissue culture cells. Chimeric control rAbs were generated from anti-myelin hybridomas in which murine variable region sequences were fused to human constant region sequences. Purified rAbs were exhaustively assayed for reactivity against myelin basic protein, proteolipid protein, and myelin oligodendrocyte glycoprotein by immunostaining of transfected cells expressing individual myelin proteins, by protein immunoblotting, and by immunostaining of human brain tissue sections. RESULTS: Whereas humanized control rAbs derived from anti-myelin hybridomas and anti-myelin monoclonal antibodies readily detected myelin antigens in multiple immunoassays, none of the rAbs derived from MS cerebrospinal fluid displayed immunoreactivity to the three myelin antigens tested. Immunocytochemical analysis of tissue sections from MS and control brain demonstrated only weak staining with a few rAbs against nuclei or cytoplasmic granules in neurons, glia, and inflammatory cells. INTERPRETATION: The oligoclonal B-cell response in MS cerebrospinal fluid is not targeted to the well-characterized myelin antigens myelin basic protein, proteolipid protein, or myelin oligodendrocyte glycoprotein.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/cerebrospinal fluid , Cell Proliferation , Immunoglobulin G/biosynthesis , Immunoglobulin G/cerebrospinal fluid , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Plasma Cells/immunology , Plasma Cells/pathology , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , Cell Line , Clone Cells , Humans , Mice , Mice, Inbred BALB C , Multiple Sclerosis/pathology , Plasma Cells/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/cerebrospinal fluid
6.
J Biol Chem ; 283(49): 34021-8, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18826953

ABSTRACT

In prion disease, direct interaction between the cellular prion protein (PrP(C)) and its misfolded disease-associated conformer PrP(Sc) is a crucial, although poorly understood step promoting the formation of nascent PrP(Sc) and prion infectivity. Recently, we hypothesized that three regions of PrP (corresponding to amino acid residues 23-33, 98-110, and 136-158) interacting specifically and robustly with PrP(Sc), likely represent peptidic components of one flank of the prion replicative interface. In this study, we created epitope-tagged mouse PrP(C) molecules in which the PrP sequences 23-33, 98-110, and 136-158 were modified. These novel PrP molecules were individually expressed in the prion-infected neuroblastoma cell line (ScN2a) and the conversion of each mutated mouse PrP(C) substrate to PrP(Sc) compared with that of the epitope-tagged wild-type mouse PrP(C). Mutations within PrP 98-110, substituting all 4 wild-type lysine residues with alanine residues, prevented conversion to PrP(Sc). Furthermore, when residues within PrP 136-140 were collectively scrambled, changed to alanines, or amino acids at positions 136, 137, and 139 individually replaced by alanine, conversion to PrP(Sc) was similarly halted. However, other PrP molecules containing mutations within regions 23-33 and 101-104 were able to readily convert to PrP(Sc). These results suggest that PrP sequence comprising residues 98-110 and 136-140 not only participates in the specific binding interaction between PrP(C) and PrP(Sc), but also in the process leading to conversion of PrP(Sc)-sequestered PrP(C) into its disease-associated form.


Subject(s)
PrPC Proteins/chemistry , PrPSc Proteins/chemistry , Alanine/chemistry , Animals , Cell Line, Tumor , Epitopes/chemistry , Flow Cytometry , Mice , Mutation , Neuroblastoma/metabolism , Peptides/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Prions/chemistry , Protein Denaturation , Protein Folding , Transfection
7.
J Neurochem ; 105(6): 2190-204, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18298665

ABSTRACT

The key event in the pathogenesis of prion diseases is the conformational conversion of the normal prion protein (PrP) (PrP(C)) into an infectious, aggregated isoform (PrP(Sc)) that has a high content of beta-sheet. Historically, a great deal of effort has been devoted to developing antibodies that specifically recognize PrP(Sc) but not PrP(C), as such antibodies would have enormous diagnostic and experimental value. A mouse monoclonal IgM antibody (designated 15B3) and three PrP motif-grafted monoclonal antibodies (referred to as IgG 19-33, 89-112, and 136-158) have been previously reported to react specifically with infectious PrP(Sc) but not PrP(C). In this study, we extend the characterization of these four antibodies by testing their ability to immunoprecipitate and immunostain infectious and non-infectious aggregates of wild-type, mutant, and recombinant PrP. We find that 15B3 as well as the motif-grafted antibodies recognize multiple types of aggregated PrP, both infectious and non-infectious, including forms found in brain, in transfected cells, and induced in vitro from purified recombinant protein. These antibodies are exquisitely selective for aggregated PrP, and do not react with soluble PrP even when present in vast excess. Our results suggest that 15B3 and the motif-grafted antibodies recognize structural features common to both infectious and non-infectious aggregates of PrP. Our study extends the utility of these antibodies for diagnostic and experimental purposes, and it provides new insight into the structural changes that accompany PrP oligomerization and prion propagation.


Subject(s)
Antibodies, Monoclonal/metabolism , PrPSc Proteins/immunology , PrPSc Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Mice, Transgenic , PrPSc Proteins/pathogenicity , Prions/chemistry , Prions/immunology , Prions/metabolism , Protein Binding/physiology , Protein Isoforms/immunology , Protein Isoforms/metabolism
8.
PLoS One ; 3(10): e3314, 2008.
Article in English | MEDLINE | ID: mdl-19390657

ABSTRACT

Transmissible spongiform encephalopathies, including variant-Creutzfeldt-Jakob disease (vCJD) in humans and bovine spongiform encephalopathies in cattle, are fatal neurodegenerative disorders characterized by protein misfolding of the host cellular prion protein (PrP(C)) to the infectious scrapie form (PrP(Sc)). However, the mechanism that exogenous PrP(Sc) infects cells and where pathologic conversion of PrP(C) to the PrP(Sc) form occurs remains uncertain. Here we report that similar to the mechanism of HIV-1 TAT-mediated peptide transduction, processed mature, full length PrP contains a conserved N-terminal cationic domain that stimulates cellular uptake by lipid raft-dependent, macropinocytosis. Inhibition of macropinocytosis by three independent means prevented cellular uptake of recombinant PrP; however, it did not affect recombinant PrP cell surface association. In addition, fusion of the cationic N-terminal PrP domain to a Cre recombinase reporter protein was sufficient to promote both cellular uptake and escape from the macropinosomes into the cytoplasm. Inhibition of macropinocytosis was sufficient to prevent conversion of PrP(C) to the pathologic PrP(Sc) form in N2a cells exposed to strain RML PrP(Sc) infected brain homogenates, suggesting that a critical determinant of PrP(C) conversion occurs following macropinocytotic internalization and not through mere membrane association. Taken together, these observations provide a cellular mechanism that exogenous pathological PrP(Sc) infects cells by lipid raft dependent, macropinocytosis.


Subject(s)
Membrane Microdomains/metabolism , Pinocytosis , PrPC Proteins/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Dermatan Sulfate/metabolism , Dose-Response Relationship, Drug , Genes, Reporter , Heparin/metabolism , Integrases/metabolism , Membrane Microdomains/pathology , Mice , Molecular Sequence Data , PrPC Proteins/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Recombination, Genetic , Time Factors
9.
J Biol Chem ; 282(50): 36341-53, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17940287

ABSTRACT

The central pathogenic event of prion disease is the conformational conversion of a host protein, PrPC, into a pathogenic isoform, PrPSc. We previously showed that the protein misfolding cyclic amplification (PMCA) technique can be used to form infectious prion molecules de novo from purified native PrPC molecules in an autocatalytic process requiring accessory polyanions (Deleault, N. R., Harris, B. T., Rees, J. R., and Supattapone, S. (2007) Proc. Natl. Acad. Sci. U. S. A. 104, 9741-9746). Here we investigated the molecular mechanism by which polyanionic molecules facilitate infectious prion formation in vitro. Ina PMCA reaction lacking PrPSc template seed, synthetic polyA RNA molecules induce hamster HaPrPC to adopt a protease-sensitive, detergent-insoluble conformation reactive against antibodies specific for PrPSc. During PMCA, labeled nucleic acids form nuclease-resistant complexes with HaPrP molecules. Strikingly, purified HaPrPC molecules subjected to PMCA selectively incorporate an approximately 1-2.5-kb subset of [32P]polyA RNA molecules from a heterogeneous mixture ranging in size from approximately 0.1 to >6 kb. Neuropathological analysis of scrapie-infected hamsters using the fluorescent dye acridine orange revealed that RNA molecules co-localize with large extracellular HaPrP aggregates. These findings suggest that polyanionic molecules such as RNA may become selectively incorporated into stable complexes with PrP molecules during the formation of native hamster prions.


Subject(s)
Amyloid/metabolism , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , RNA/metabolism , Scrapie/metabolism , Amyloid/chemistry , Animals , Catalysis , Cricetinae , Cricetulus , Female , Male , Polyelectrolytes , Polymers/chemistry , Polymers/metabolism , PrPC Proteins/chemistry , PrPSc Proteins/chemistry , RNA/chemistry , Ribonucleases/chemistry , Scrapie/pathology
10.
J Biol Chem ; 282(49): 35878-86, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17923484

ABSTRACT

Diagnosis of transmissible spongiform encephalopathy (TSE) disease in humans and ruminants relies on the detection in post-mortem brain tissue of the protease-resistant form of the host glycoprotein PrP. The presence of this abnormal isoform (PrP(Sc)) in tissues is taken as indicative of the presence of TSE infectivity. Here we demonstrate conclusively that high titers of TSE infectivity can be present in brain tissue of animals that show clinical and vacuolar signs of TSE disease but contain low or undetectable levels of PrP(Sc). This work questions the correlation between PrP(Sc) level and the titer of infectivity and shows that tissues containing little or no proteinase K-resistant PrP can be infectious and harbor high titers of TSE infectivity. Reliance on protease-resistant PrP(Sc) as a sole measure of infectivity may therefore in some instances significantly underestimate biological properties of diagnostic samples, thereby undermining efforts to contain and eradicate TSEs.


Subject(s)
Brain Chemistry , Endopeptidase K/chemistry , PrPSc Proteins/analysis , Prion Diseases/diagnosis , Animals , Brain/metabolism , Brain/pathology , Cricetinae , Mice , Mice, Transgenic , PrPSc Proteins/metabolism , PrPSc Proteins/pathogenicity , Prion Diseases/metabolism , Prion Diseases/transmission , Ruminants/metabolism
11.
J Virol ; 81(15): 8315-24, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17522220

ABSTRACT

Human metapneumovirus (hMPV) is a recently discovered paramyxovirus that is a major cause of lower-respiratory-tract disease. hMPV is associated with more severe disease in infants and persons with underlying medical conditions. Animal studies have shown that the hMPV fusion (F) protein alone is capable of inducing protective immunity. Here, we report the use of phage display technology to generate a fully human monoclonal antibody fragment (Fab) with biological activity against hMPV. Phage antibody libraries prepared from human donor tissues were selected against recombinant hMPV F protein with multiple rounds of panning. Recombinant Fabs then were expressed in bacteria, and supernatants were screened by enzyme-linked immunosorbent assay and immunofluorescent assays. A number of Fabs that bound to hMPV F were isolated, and several of these exhibited neutralizing activity in vitro. Fab DS7 neutralized the parent strain of hMPV with a 60% plaque reduction activity of 1.1 mug/ml and bound to hMPV F with an affinity of 9.8 x10(-10) M, as measured by surface plasmon resonance. To test the in vivo activity of Fab DS7, groups of cotton rats were infected with hMPV and given Fab intranasally 3 days after infection. Nasal turbinates and lungs were harvested on day 4 postinfection and virus titers determined. Animals treated with Fab DS7 exhibited a >1,500-fold reduction in viral titer in the lungs, with a modest 4-fold reduction in the nasal tissues. There was a dose-response relationship between the dose of DS7 and virus titer. Human Fab DS7 may have prophylactic or therapeutic potential against severe hMPV infection.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin Fab Fragments/immunology , Metapneumovirus/immunology , Paramyxoviridae Infections/therapy , Recombinant Proteins/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal/genetics , Cell Line , Epitopes , Humans , Immunoglobulin Fab Fragments/genetics , Infant , Metapneumovirus/genetics , Neutralization Tests , Peptide Library , Rats , Recombinant Proteins/genetics , Sigmodontinae , Viral Fusion Proteins/genetics , Viral Load
12.
J Biol Chem ; 282(10): 7465-71, 2007 Mar 09.
Article in English | MEDLINE | ID: mdl-17218310

ABSTRACT

Direct interaction between endogenous cellular prion protein (PrP(C)) and misfolded, disease-associated (PrP(Sc)) conformers is a key event in prion propagation, which precedes templated conversion of PrP(C) into nascent PrP(Sc) and prion infectivity. Although almost none of the molecular details of this pivotal process are understood, the persistence of individual prion strains suggests that assembly of the prion replicative complex is mechanistically precise. To systematically map defined regions of PrP(C) sequence that bind tightly to PrP(Sc), we have generated a comprehensive panel of over 45 motif-grafted antibodies containing overlapping peptide grafts collectively spanning PrP residues 19-231. Grafted antibody binding experiments, performed under stringent conditions, clearly identified only three distinct and independent high affinity PrP(Sc) recognition motifs. The first of these binding motifs lies at the very N-terminal region of the mature PrP molecule within PrP-(23-33); the second motif lies within PrP-(98-110); and the third is contained within PrP-(136-158). Mutational analyses of these PrP(Sc)-binding regions revealed that reactivity of the 23-33 and 98-110 segments are largely dependent upon the presence of multiple positively charged amino acid residues. These studies yield new insight into critical peptidic components composing one side of the prion replicative interface.


Subject(s)
PrPC Proteins/chemistry , PrPSc Proteins/chemistry , Protein Interaction Mapping/methods , Binding Sites , Humans , Point Mutation , Protein Conformation
13.
J Mol Biol ; 363(1): 75-97, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16962610

ABSTRACT

Bovine spongiform encephalopathy (BSE) is a fatal neurodegenerative prion disease affecting cattle that is transmissible to humans, manifesting as a variant of Creutzfeldt-Jakob disease (vCJD) likely following the consumption of meat contaminated with BSE prions. High-affinity antibodies are a prerequisite for the development of simple, highly sensitive and non-invasive diagnostic tests that are able to detect even small amounts of the disease-associated PrP conformer (PrP(Sc)). We describe here the affinity maturation of a single-chain Fv antibody fragment with a binding affinity of 1 pM to a peptide derived from the unstructured region of bovine PrP (BoPrP (90-105)). This is the tightest peptide-binding antibody reported to date and may find useful application in diagnostics, especially when PrP(Sc) is pretreated by denaturation and/or proteolysis for peptide-like presentation. Several rounds of directed evolution and off-rate selection with ribosome display were performed using an antibody library generated from a single PrP binder with error-prone PCR and DNA-shuffling. As the correct determinations of affinities in this range are not straightforward, competition biosensor techniques and KinExA methods were both applied and compared. Structural interpretation of the affinity improvement was performed based on the crystal structure of the original prion binder in complex with the BoPrP (95-104) peptide by modeling the corresponding mutations.


Subject(s)
Directed Molecular Evolution , Immunoglobulin Fragments/chemistry , Immunoglobulin Variable Region/chemistry , Prions/immunology , Amino Acid Sequence , Amino Acid Substitution/genetics , Brain/immunology , Brain/metabolism , Brain Chemistry , Crystallography, X-Ray , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Molecular Sequence Data , Protein Binding
14.
J Biol Chem ; 281(22): 15536-45, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16569635

ABSTRACT

The coexistence of multiple strains or subtypes of the disease-related isoform of prion protein (PrP) in natural isolates, together with the observed conformational heterogeneity of PrP amyloid fibrils generated in vitro, indicates the importance of probing the conformation of single particles within heterogeneous samples. Using an array of PrP-specific antibodies, we report the development of a novel immunoconformational assay. Uniquely, application of this new technology allows the conformation of multimeric PrP within a single fibril or particle to be probed without pretreatment of the sample with proteinase K. Using amyloid fibrils prepared from full-length recombinant PrP, we demonstrated the utility of this assay to define (i) PrP regions that are surface-exposed or buried, (ii) the susceptibility of defined PrP regions to GdnHCl-induced denaturation, and (iii) the conformational heterogeneity of PrP fibrils as measured for either the entire fibrillar population or for individual fibrils. Specifically, PrP regions 159-174 and 224-230 were shown to be buried and were the most resistant to denaturation. The 132-156 segment of PrP was found to be cryptic under native conditions and solvent-exposed under partially denaturing conditions, whereas the region 95-105 was solvent-accessible regardless of the solvent conditions. Remarkably, a subfraction of fibrils showed immunoreactivity to PrPSc-specific antibodies designated as IgGs 89-112 and 136-158. The immunoreactivity of the conformational epitopes was reduced upon exposure to partially denaturing conditions. Unexpectedly, PrPSc -specific antibodies revealed conformational polymorphisms even within individual fibrils. Our studies provide valuable new insight into fibrillar substructure and offer a new tool for probing the conformation of single PrP fibrils.


Subject(s)
Amyloid/chemistry , Prions/chemistry , Protein Precursors/chemistry , Amyloid/genetics , Amyloid/immunology , Animals , Epitopes/chemistry , Immunochemistry , In Vitro Techniques , Mice , Microscopy, Fluorescence , Prion Proteins , Prions/genetics , Prions/immunology , Protein Conformation , Protein Denaturation , Protein Precursors/genetics , Protein Precursors/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology
15.
J Biol Chem ; 280(29): 26873-9, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15917229

ABSTRACT

Little is currently known about the biochemical mechanism by which induced prion protein (PrP) conformational change occurs during mammalian prion propagation. In this study, we describe the reconstitution of PrPres amplification in vitro using partially purified and synthetic components. Overnight incubation of purified PrP27-30 and PrPC molecules at a molar ratio of 1:250 yielded approximately 2-fold baseline PrPres amplification. Addition of various polyanionic molecules increased the level of PrPres amplification to approximately 10-fold overall. Polyanionic compounds that stimulated purified PrPres amplification to varying degrees included synthetic, homopolymeric nucleic acids such as poly(A) and poly(dT), as well as non-nucleic acid polyanions, such as heparan sulfate proteoglycan. Size fractionation experiments showed that synthetic poly(A) polymers must be >0.2 kb in length to stimulate purified PrPres amplification. Thus, one possible set of minimal components for efficient conversion of PrP molecules in vitro may be surprisingly simple, consisting of PrP27-30, PrPC, and a stimulatory polyanionic compound.


Subject(s)
Peptide Fragments/metabolism , Peptide Hydrolases/metabolism , Polymers/pharmacology , PrPC Proteins/metabolism , Animals , Cricetinae , Heparin/analogs & derivatives , Heparin/pharmacology , Humans , Poly A/pharmacology , Poly T/pharmacology , Polyelectrolytes , Protein Conformation , Proteoglycans/pharmacology
16.
Proc Natl Acad Sci U S A ; 101(28): 10404-9, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15240877

ABSTRACT

Prion diseases are closely associated with the conversion of the cellular prion protein (PrPC) to an abnormal conformer (PrPSc) [Prusiner, S. B. (1998) Proc. Natl. Acad. Sci. USA 95, 13363-13383]. Monoclonal antibodies that bind epitopes comprising residues 96-104 and 133-158 of PrPC potently inhibit this process, presumably by preventing heterodimeric association of PrPC and PrPSc, and suggest that these regions of PrPC may be critical components of the PrPC-PrPSc replicative interface. We reasoned that transplanting PrP sequence corresponding to these regions into a suitable carrier molecule, such as an antibody, could impart specific recognition of disease-associated forms of PrP. To test this hypothesis, polypeptides containing PrP sequence between residues 89-112 or 136-158 were used to replace the extended heavy chain complementarity-determining region 3 of an IgG antibody specific for the envelope glycoprotein of HIV-1. Herein the resulting engineered PrP-IgGs are shown to bind specifically to infective fractions of PrP in mouse, human, and hamster prion-infected tissues, but not to PrPC, other cellular components, or the HIV-1 envelope. PrPSc reactivity was abolished when the sequence of the PrP 89-112 and 136-158 grafts was mutated, scrambled, or N-terminally truncated. Our findings suggest that residues within the 89-112 and 136-158 segments of PrPC are key components of one face of the PrPC-PrPSc complex. PrPSc-specific antibodies produced by the approach described may find widespread application in the study of prion biology and replication and in the detection of infectious prions in human and animal materials.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity , PrPC Proteins/immunology , PrPSc Proteins/immunology , Scrapie/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Creutzfeldt-Jakob Syndrome/immunology , Cricetinae , Epitopes/genetics , Epitopes/immunology , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Molecular Sequence Data , PrPC Proteins/genetics , PrPSc Proteins/genetics
17.
J Immunol ; 173(1): 649-56, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15210828

ABSTRACT

Increased amounts of intrathecally synthesized IgG and oligoclonal bands have long been recognized as a hallmark of multiple sclerosis (MS). B cells and plasma cells are components of the inflammatory infiltrates in both active and chronic MS lesions, and increased numbers of these cells are present in MS cerebrospinal fluid (CSF). Single-cell RT-PCR was used to analyze both the CD19+ B cell and CD138+ plasma cell populations in CSF of two patients with clinically definite MS and of one MS patient whose CSF was obtained after a clinically isolated syndrome, but before the second episode. Sequence analysis of amplified IgG V region sequences identified the rearranged germline segments, extent of somatic mutation, and clonal relationships within and between the two cell populations in the three MS patients. Expanded B cell and plasma cell clones were detected in each MS CSF and in all three patients the CD138+ IgG repertoire was more restricted. However, little if any significant sequence overlap was observed between the CD19+ and CD138+ repertoires of each donor. Detection of plasma cell clones by single-cell PCR will facilitate the in vitro production of recombinant Abs useful in identifying disease-relevant Ags.


Subject(s)
Antigens, CD19/analysis , Immunoglobulin Variable Region/chemistry , Membrane Glycoproteins/analysis , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Proteoglycans/analysis , Adult , Amino Acid Sequence , B-Lymphocytes/immunology , Female , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Middle Aged , Molecular Sequence Data , Plasma Cells/immunology , Syndecan-1 , Syndecans
18.
J Biol Chem ; 279(21): 21948-56, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15026410

ABSTRACT

Previous studies using post-mortem human brain extracts demonstrated that PrP in Creutzfeldt-Jakob disease (CJD) brains is cleaved by a cellular protease to generate a C-terminal fragment, referred to as C2, which has the same molecular weight as PrP-(27-30), the protease-resistant core of PrP(Sc) (1). The role of this endoproteolytic cleavage of PrP in prion pathogenesis and the identity of the cellular protease responsible for production of the C2 cleavage product has not been explored. To address these issues we have taken a combination of pharmacological and genetic approaches using persistently infected scrapie mouse brain (SMB) cells. We confirm that production of C2 is the predominant cleavage event of PrP(Sc) in the brains of scrapie-infected mice and that SMB cells faithfully recapitulate the diverse intracellular proteolytic processing events of PrP(Sc) and PrP(C) observed in vivo. While increases in intracellular calcium (Ca(2+)) levels in prion-infected cell cultures stimulate the production of the PrP(Sc) cleavage product, pharmacological inhibitors of calpains and overexpression of the endogenous calpain inhibitor, calpastatin, prevent the production of C2. In contrast, inhibitors of lysosomal proteases, caspases, and the proteasome have no effect on C2 production in SMB cells. Calpain inhibition also prevents the accumulation of PrP(Sc) in SMB and persistently infected ScN2A cells, whereas bioassay of inhibitor-treated cell cultures demonstrates that calpain inhibition results in reduced prion titers compared with control-treated cultures assessed in parallel. Our observations suggest that calpain-mediated endoproteolytic cleavage of PrP(Sc) may be an important event in prion propagation.


Subject(s)
Calpain/metabolism , PrPSc Proteins/metabolism , Prions/chemistry , Scrapie/metabolism , Animals , Biological Assay , Brain/metabolism , Calcium/chemistry , Calcium-Binding Proteins/pharmacology , Calpain/chemistry , Cell Division , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Ionophores/pharmacology , Kinetics , Mice , PrPC Proteins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Time Factors
19.
Science ; 303(5663): 1514-6, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-14752167

ABSTRACT

Neuronal death is a prominent, but poorly understood, pathological hallmark of prion disease. Notably, in the absence of the cellular prion protein (PrPC), the disease-associated isoform, PrPSc, appears not to be intrinsically neurotoxic, suggesting that PrPC itself may participate directly in the prion neurodegenerative cascade. Here, cross-linking PrPC in vivo with specific monoclonal antibodies was found to trigger rapid and extensive apoptosis in hippocampal and cerebellar neurons. These findings suggest that PrPC functions in the control of neuronal survival and provides a model to explore whether cross-linking of PrPC by oligomeric PrPSc can promote neuronal loss during prion infection.


Subject(s)
Antibodies, Monoclonal/metabolism , Apoptosis , Cerebellum/cytology , Hippocampus/cytology , Neurons/physiology , PrPC Proteins/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Survival , Complement Activation , Dimerization , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Neural Cell Adhesion Molecules/immunology , Neural Cell Adhesion Molecules/metabolism , PrPC Proteins/chemistry , PrPC Proteins/immunology , Recombinant Proteins/metabolism , Signal Transduction
20.
J Immunol ; 171(5): 2725-33, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12928426

ABSTRACT

Single-cell RT-PCR was used to sample CD19(+) B cell repertoires in cerebrospinal fluid (CSF) of patients with multiple sclerosis (MS) or viral meningitis. Analysis of amplified Ab H and L chain products served to identify the rearranged germline segment and J segment, and to determine the degree of homology for the H and L chain sequence of individual B cells. The B cell repertoire of viral meningitis CSF was predominantly polyclonal, whereas B cell clonal expansion was a prominent feature of the IgG repertoire in three of four MS patients. Two dominant clonal populations in one MS CSF accounted for approximately 70% of the IgG H chain V regions sequenced, while the corresponding IgM repertoires were more heterogeneous. One clonal B cell population revealed multiple L chain rearrangements, raising the possibility of a role for receptor editing in shaping the B cell response in some MS patients. The most immediate implications of identifying rearranged Ig sequences in MS B cells is the potential to accurately recreate recombinant Abs from these overrepresented H and L chains that can be used to discover the relevant Ag(s) in MS.


Subject(s)
B-Lymphocyte Subsets/immunology , Lymphocyte Activation/immunology , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Acute Disease , Adult , Amino Acid Sequence , Antigens, CD19/biosynthesis , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , Cell Separation , Clone Cells , Female , Flow Cytometry , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Gene Rearrangement, B-Lymphocyte, Light Chain , Humans , Immunoglobulin Heavy Chains/cerebrospinal fluid , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/cerebrospinal fluid , Immunoglobulin Variable Region/genetics , Lymphocyte Activation/genetics , Male , Meningitis, Viral/cerebrospinal fluid , Meningitis, Viral/genetics , Meningitis, Viral/immunology , Middle Aged , Molecular Sequence Data , Multiple Sclerosis/genetics , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...