Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Front Immunol ; 12: 706757, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335620

RESUMEN

Three clinically relevant ebolaviruses - Ebola (EBOV), Bundibugyo (BDBV), and Sudan (SUDV) viruses, are responsible for severe disease and occasional deadly outbreaks in Africa. The largest Ebola virus disease (EVD) epidemic to date in 2013-2016 in West Africa highlighted the urgent need for countermeasures, leading to the development and FDA approval of the Ebola virus vaccine rVSV-ZEBOV (Ervebo®) in 2020 and two monoclonal antibody (mAb)-based therapeutics (Inmazeb® [atoltivimab, maftivimab, and odesivimab-ebgn] and Ebanga® (ansuvimab-zykl) in 2020. The humoral response plays an indispensable role in ebolavirus immunity, based on studies of mAbs isolated from the antibody genes in peripheral blood circulating ebolavirus-specific human memory B cells. However, antibodies in the body are not secreted by circulating memory B cells in the blood but rather principally by plasma cells in the bone marrow. Little is known about the protective polyclonal antibody responses in convalescent plasma. Here we exploited both single-cell antibody gene sequencing and proteomic sequencing approaches to assess the composition of the ebolavirus glycoprotein (GP)-reactive antibody repertoire in the plasma of an EVD survivor. We first identified 1,512 GP-specific mAb variable gene sequences from single cells in the memory B cell compartment. Using mass spectrometric analysis of the corresponding GP-specific plasma IgG, we found that only a portion of the large B cell antibody repertoire was represented in the plasma. Molecular and functional analysis of proteomics-identified mAbs revealed recognition of epitopes in three major antigenic sites - the GP head domain, the glycan cap, and the base region, with a high prevalence of neutralizing and protective mAb specificities that targeted the base and glycan cap regions on the GP. Polyclonal plasma antibodies from the survivor reacted broadly to EBOV, BDBV, and SUDV GP, while reactivity of the potently neutralizing mAbs we identified was limited mostly to the homologous EBOV GP. Together these results reveal a restricted diversity of neutralizing humoral response in which mAbs targeting two antigenic sites on GP - glycan cap and base - play a principal role in plasma-antibody-mediated protective immunity against EVD.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Ebolavirus/inmunología , Glicoproteínas de Membrana/inmunología , Adulto , Fiebre Hemorrágica Ebola/inmunología , Humanos , Masculino , Proteómica
2.
Toxins (Basel) ; 12(4)2020 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-32235318

RESUMEN

PB10 IgG1, a monoclonal antibody (MAb) directed against an immunodominant epitope on the enzymatic subunit (RTA) of ricin toxin (RT), has been shown to passively protect mice and non-human primates from an aerosolized lethal-dose RT challenge. However, it was recently demonstrated that the therapeutic efficacy of PB10 IgG1 is significantly improved when co-administered with a second MAb, SylH3, targeting RT's binding subunit (RTB). Here we report that the PB10/SylH3 cocktail is also superior to PB10 alone when used as a pre-exposure prophylactic (PrEP) in a mouse model of intranasal RT challenge. The benefit of the PB10/SylH3 cocktail prompted us to engineer a humanized IgG1 version of SylH3 (huSylH3). The huPB10/huSylH3 cocktail proved highly efficacious in the mouse model, thereby opening the door to future testing in non-human primates.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Antídotos/farmacología , Enfermedades Pulmonares/prevención & control , Ricina/antagonistas & inhibidores , Administración por Inhalación , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Antídotos/administración & dosificación , Chlorocebus aethiops , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Enfermedades Pulmonares/inducido químicamente , Ratones Endogámicos BALB C , Profilaxis Pre-Exposición , Ricina/inmunología , Células Vero
3.
Anal Chem ; 92(1): 766-773, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31769659

RESUMEN

One challenge associated with the discovery and development of monoclonal antibody (mAb) therapeutics is the determination of heavy chain and light chain pairing. Advances in MS instrumentation and MS/MS methods have greatly enhanced capabilities for the analysis of large intact proteins yielding much more detailed and accurate proteoform characterization. Consequently, direct interrogation of intact antibodies or F(ab')2 and Fab fragments has the potential to significantly streamline therapeutic mAb discovery processes. Here, we demonstrate for the first time the ability to efficiently cleave disulfide bonds linking heavy and light chains of mAbs using electron capture dissociation (ECD) and 157 nm ultraviolet photodissociation (UVPD). The combination of intact mAb, Fab, or F(ab')2 mass, intact LC and Fd masses, and CDR3 sequence coverage enabled determination of heavy chain and light chain pairing from a single experiment and experimental condition. These results demonstrate the potential of top-down and middle-down proteomics to significantly streamline therapeutic antibody discovery.


Asunto(s)
Anticuerpos Monoclonales/química , Secuencia de Aminoácidos , Antineoplásicos Inmunológicos/química , Fragmentos Fab de Inmunoglobulinas/análisis , Cadenas Pesadas de Inmunoglobulina/análisis , Cadenas Ligeras de Inmunoglobulina/análisis , Espectrometría de Masas , Fotólisis , Trastuzumab/química , Rayos Ultravioleta
4.
Analyst ; 143(16): 3907-3917, 2018 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-30039812

RESUMEN

Proteolytic digestion is an important step in characterizing protein sequences and post-translational modifications (PTMs) using mass spectrometry (MS). This study uses pepsin- or trypsin-containing spin membranes for rapid digestion of single proteins or simple protein mixtures prior to ultrahigh-resolution Orbitrap MS analysis. Centrifugation of 100 µL of pretreated protein solutions through the functionalized membranes requires less than 1 min and conveniently digests proteins into large peptides that aid in confirming specific protein sequence variations and PTMs. Peptic and tryptic peptides from spin digestion of apomyoglobin and four commercial monoclonal antibodies (mAbs) typically cover 100% of the protein sequences in direct infusion MS analysis. Increasing the spin rate leads to a higher fraction of large peptic peptides for apomyoglobin, and MS analysis of peptic and tryptic peptides reveals mAb PTMs such as N-terminal pyroglutamate formation, C-terminal lysine clipping and glycosylation. Relative to overnight in-solution digestion of mAbs, spin digestion yields higher sequence coverages. Spin-membrane digestion followed by infusion MS readily differentiates a mAb to the Ebola virus from a related antibody that differs by addition of a single amino acid.


Asunto(s)
Péptidos/química , Procesamiento Proteico-Postraduccional , Proteolisis , Secuencia de Aminoácidos , Anticuerpos Monoclonales/química , Apoproteínas/química , Espectrometría de Masas , Mioglobina/química , Pepsina A/química , Tripsina/química
5.
Sci Data ; 4: 170090, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28696408

RESUMEN

Venoms are a rich source for the discovery of molecules with biotechnological applications, but their analysis is challenging even for state-of-the-art proteomics. Here we report on a large-scale proteomic assessment of the venom of Loxosceles intermedia, the so-called brown spider. Venom was extracted from 200 spiders and fractioned into two aliquots relative to a 10 kDa cutoff mass. Each of these was further fractioned and digested with trypsin (4 h), trypsin (18 h), pepsin (18 h), and chymotrypsin (18 h), then analyzed by MudPIT on an LTQ-Orbitrap XL ETD mass spectrometer fragmenting precursors by CID, HCD, and ETD. Aliquots of undigested samples were also analyzed. Our experimental design allowed us to apply spectral networks, thus enabling us to obtain meta-contig assemblies, and consequently de novo sequencing of practically complete proteins, culminating in a deep proteome assessment of the venom. Data are available via ProteomeXchange, with identifier PXD005523.


Asunto(s)
Proteoma , Venenos de Araña/química , Arañas , Animales , Espectrometría de Masas , Péptido Hidrolasas , Proteómica
6.
J Proteome Res ; 16(1): 45-54, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27779884

RESUMEN

One direct route for the discovery of therapeutic human monoclonal antibodies (mAbs) involves the isolation of peripheral B cells from survivors/sero-positive individuals after exposure to an infectious reagent or disease etiology, followed by single-cell sequencing or hybridoma generation. Peripheral B cells, however, are not always easy to obtain and represent only a small percentage of the total B-cell population across all bodily tissues. Although it has been demonstrated that tandem mass spectrometry (MS/MS) techniques can interrogate the full polyclonal antibody (pAb) response to an antigen in vivo, all current approaches identify MS/MS spectra against databases derived from genetic sequencing of B cells from the same patient. In this proof-of-concept study, we demonstrate the feasibility of a novel MS/MS antibody discovery approach in which only serum antibodies are required without the need for sequencing of genetic material. Peripheral pAbs from a cytomegalovirus-exposed individual were purified by glycoprotein B antigen affinity and de novo sequenced from MS/MS data. Purely MS-derived mAbs were then manufactured in mammalian cells to validate potency via antigen-binding ELISA. Interestingly, we found that these mAbs accounted for 1 to 2% of total donor IgG but were not detected in parallel sequencing of memory B cells from the same patient.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Antivirales/química , Linfocitos B/inmunología , Infecciones por Citomegalovirus/inmunología , Inmunoglobulina G/química , Análisis de Secuencia de Proteína , Secuencia de Aminoácidos , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/aislamiento & purificación , Formación de Anticuerpos , Células Productoras de Anticuerpos/citología , Células Productoras de Anticuerpos/inmunología , Linfocitos B/virología , Cromatografía de Afinidad/métodos , Citomegalovirus/crecimiento & desarrollo , Infecciones por Citomegalovirus/sangre , Infecciones por Citomegalovirus/virología , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Sueros Inmunes/química , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/aislamiento & purificación , Espectrometría de Masas en Tándem , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología
7.
J Comput Biol ; 22(5): 353-66, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25423621

RESUMEN

Tandem mass (MS/MS) spectrometry has become the method of choice for protein identification and has launched a quest for the identification of every translated protein and peptide. However, computational developments have lagged behind the pace of modern data acquisition protocols and have become a major bottleneck in proteomics analysis of complex samples. As it stands today, attempts to identify MS/MS spectra against large databases (e.g., the human microbiome or 6-frame translation of the human genome) face a search space that is 10-100 times larger than the human proteome, where it becomes increasingly challenging to separate between true and false peptide matches. As a result, the sensitivity of current state-of-the-art database search methods drops by nearly 38% to such low identification rates that almost 90% of all MS/MS spectra are left as unidentified. We address this problem by extending the generating function approach to rigorously compute the joint spectral probability of multiple spectra being matched to peptides with overlapping sequences, thus enabling the confident assignment of higher significance to overlapping peptide-spectrum matches (PSMs). We find that these joint spectral probabilities can be several orders of magnitude more significant than individual PSMs, even in the ideal case when perfect separation between signal and noise peaks could be achieved per individual MS/MS spectrum. After benchmarking this approach on a typical lysate MS/MS dataset, we show that the proposed intersecting spectral probabilities for spectra from overlapping peptides improve peptide identification by 30-62%.


Asunto(s)
Modelos Estadísticos , Péptidos/análisis , Proteómica/estadística & datos numéricos , Programas Informáticos , Espectrometría de Masas en Tándem/estadística & datos numéricos , Algoritmos , Simulación por Computador , Bases de Datos de Proteínas , Humanos , Biblioteca de Péptidos , Proteómica/métodos
8.
Mol Cell Proteomics ; 12(12): 3812-23, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24043425

RESUMEN

We report the use of neutron-encoded (NeuCode) stable isotope labeling of amino acids in cell culture for the purpose of C-terminal product ion annotation. Two NeuCode labeling isotopologues of lysine, (13)C6(15)N2 and (2)H8, which differ by 36 mDa, were metabolically embedded in a sample proteome, and the resultant labeled proteins were combined, digested, and analyzed via liquid chromatography and mass spectrometry. With MS/MS scan resolving powers of ~50,000 or higher, product ions containing the C terminus (i.e. lysine) appear as a doublet spaced by exactly 36 mDa, whereas N-terminal fragments exist as a single m/z peak. Through theory and experiment, we demonstrate that over 90% of all y-type product ions have detectable doublets. We report on an algorithm that can extract these neutron signatures with high sensitivity and specificity. In other words, of 15,503 y-type product ion peaks, the y-type ion identification algorithm correctly identified 14,552 (93.2%) based on detection of the NeuCode doublet; 6.8% were misclassified (i.e. other ion types that were assigned as y-type products). Searching NeuCode labeled yeast with PepNovo(+) resulted in a 34% increase in correct de novo identifications relative to searching through MS/MS only. We use this tool to simplify spectra prior to database searching, to sort unmatched tandem mass spectra for spectral richness, for correlation of co-fragmented ions to their parent precursor, and for de novo sequence identification.


Asunto(s)
Algoritmos , Marcaje Isotópico/métodos , Anotación de Secuencia Molecular/métodos , Neutrones , Proteoma/aislamiento & purificación , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Secuencia de Aminoácidos , Cromatografía Liquida , Expresión Génica , Iones , Lisina/metabolismo , Datos de Secuencia Molecular , Peso Molecular , Proteoma/genética , Proteoma/metabolismo , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Espectrometría de Masas en Tándem
9.
J Proteome Res ; 12(6): 2846-57, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23679345

RESUMEN

Full-length de novo sequencing of unknown proteins remains a challenging open problem. Traditional methods that sequence spectra individually are limited by short peptide length, incomplete peptide fragmentation, and ambiguous de novo interpretations. We address these issues by determining consensus sequences for assembled tandem mass (MS/MS) spectra from overlapping peptides (e.g., by using multiple enzymatic digests). We have combined electron-transfer dissociation (ETD) with collision-induced dissociation (CID) and higher-energy collision-induced dissociation (HCD) fragmentation methods to boost interpretation of long, highly charged peptides and take advantage of corroborating b/y/c/z ions in CID/HCD/ETD. Using these strategies, we show that triplet CID/HCD/ETD MS/MS spectra from overlapping peptides yield de novo sequences of average length 70 AA and as long as 200 AA at up to 99% sequencing accuracy.


Asunto(s)
Algoritmos , Fragmentos de Péptidos/aislamiento & purificación , Proteínas/química , Análisis de Secuencia de Proteína/métodos , Espectrometría de Masas en Tándem/métodos , Animales , Armoracia/química , Escherichia coli/química , Caballos , Humanos , Ratones , Proteolisis , Sensibilidad y Especificidad
10.
Mol Cell Proteomics ; 11(10): 1084-96, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22798278

RESUMEN

Full-length de novo sequencing from tandem mass (MS/MS) spectra of unknown proteins such as antibodies or proteins from organisms with unsequenced genomes remains a challenging open problem. Conventional algorithms designed to individually sequence each MS/MS spectrum are limited by incomplete peptide fragmentation or low signal to noise ratios and tend to result in short de novo sequences at low sequencing accuracy. Our shotgun protein sequencing (SPS) approach was developed to ameliorate these limitations by first finding groups of unidentified spectra from the same peptides (contigs) and then deriving a consensus de novo sequence for each assembled set of spectra (contig sequences). But whereas SPS enables much more accurate reconstruction of de novo sequences longer than can be recovered from individual MS/MS spectra, it still requires error-tolerant matching to homologous proteins to group smaller contig sequences into full-length protein sequences, thus limiting its effectiveness on sequences from poorly annotated proteins. Using low and high resolution CID and high resolution HCD MS/MS spectra, we address this limitation with a Meta-SPS algorithm designed to overlap and further assemble SPS contigs into Meta-SPS de novo contig sequences extending as long as 100 amino acids at over 97% accuracy without requiring any knowledge of homologous protein sequences. We demonstrate Meta-SPS using distinct MS/MS data sets obtained with separate enzymatic digestions and discuss how the remaining de novo sequencing limitations relate to MS/MS acquisition settings.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Fragmentos de Péptidos/análisis , Proteínas/análisis , Análisis de Secuencia de Proteína/métodos , Secuencia de Aminoácidos , Animales , Armoracia/genética , Bovinos , Biología Computacional/estadística & datos numéricos , Escherichia coli/genética , Caballos/genética , Humanos , Ratones , Datos de Secuencia Molecular , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Análisis de Secuencia de Proteína/estadística & datos numéricos , Espectrometría de Masas en Tándem/normas
11.
Mol Cell Proteomics ; 11(9): 550-7, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22595789

RESUMEN

The high-throughput nature of proteomics mass spectrometry is enabled by a productive combination of data acquisition protocols and the computational tools used to interpret the resulting spectra. One of the key components in mainstream protocols is the generation of tandem mass (MS/MS) spectra by peptide fragmentation using collision induced dissociation, the approach currently used in the large majority of proteomics experiments to routinely identify hundreds to thousands of proteins from single mass spectrometry runs. Complementary to these, alternative peptide fragmentation methods such as electron capture/transfer dissociation and higher-energy collision dissociation have consistently achieved significant improvements in the identification of certain classes of peptides, proteins, and post-translational modifications. Recognizing these advantages, mass spectrometry instruments now conveniently support fine-tuned methods that automatically alternate between peptide fragmentation modes for either different types of peptides or for acquisition of multiple MS/MS spectra from each peptide. But although these developments have the potential to substantially improve peptide identification, their routine application requires corresponding adjustments to the software tools and procedures used for automated downstream processing. This review discusses the computational implications of alternative and alternate modes of MS/MS peptide fragmentation and addresses some practical aspects of using such protocols for identification of peptides and post-translational modifications.


Asunto(s)
Fragmentos de Péptidos/análisis , Espectrometría de Masas en Tándem , Secuencia de Aminoácidos , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Péptidos/análisis , Péptidos/química , Procesamiento Proteico-Postraduccional , Proteómica , Programas Informáticos
12.
Mol Biosyst ; 8(10): 2535-44, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22610447

RESUMEN

High-throughput proteomics is made possible by a combination of modern mass spectrometry instruments capable of generating many millions of tandem mass (MS(2)) spectra on a daily basis and the increasingly sophisticated associated software for their automated identification. Despite the growing accumulation of collections of identified spectra and the regular generation of MS(2) data from related peptides, the mainstream approach for peptide identification is still the nearly two decades old approach of matching one MS(2) spectrum at a time against a database of protein sequences. Moreover, database search tools overwhelmingly continue to require that users guess in advance a small set of 4-6 post-translational modifications that may be present in their data in order to avoid incurring substantial false positive and negative rates. The spectral networks paradigm for analysis of MS(2) spectra differs from the mainstream database search paradigm in three fundamental ways. First, spectral networks are based on matching spectra against other spectra instead of against protein sequences. Second, spectral networks find spectra from related peptides even before considering their possible identifications. Third, spectral networks determine consensus identifications from sets of spectra from related peptides instead of separately attempting to identify one spectrum at a time. Even though spectral networks algorithms are still in their infancy, they have already delivered the longest and most accurate de novo sequences to date, revealed a new route for the discovery of unexpected post-translational modifications and highly-modified peptides, enabled automated sequencing of cyclic non-ribosomal peptides with unknown amino acids and are now defining a novel approach for mapping the entire molecular output of biological systems that is suitable for analysis with tandem mass spectrometry. Here we review the current state of spectral networks algorithms and discuss possible future directions for automated interpretation of spectra from any class of molecules.


Asunto(s)
Oligonucleótidos/química , Péptidos Cíclicos/química , Proteómica/métodos , Programas Informáticos , Espectrometría de Masas en Tándem/métodos , Algoritmos , Secuencia de Aminoácidos , Automatización de Laboratorios , Bacillus subtilis/química , Catarata/metabolismo , Bases de Datos de Proteínas , Humanos , Cristalino/química , Datos de Secuencia Molecular , Oligonucleótidos/análisis , Péptidos Cíclicos/análisis , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA