Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Proteomics ; 23(5): 100753, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38527648

RESUMO

Bacterial or viral antigens can contain subdominant protein regions that elicit weak antibody responses upon vaccination or infection although there is accumulating evidence that antibody responses against subdominant regions can enhance the protective immune response. One proposed mechanism for subdominant protein regions is the binding of host proteins that prevent antibody production against epitopes hidden within the protein binding interfaces. Here, we used affinity purification combined with quantitative mass spectrometry (AP-MS) to examine the level of competition between antigen-specific antibodies and host-pathogen protein interaction networks using the M1 protein from Streptococcus pyogenes as a model system. As most humans have circulating antibodies against the M1 protein, we first used AP-MS to show that the M1 protein interspecies protein network formed with human plasma proteins is largely conserved in naïve mice. Immunizing mice with the M1 protein generated a time-dependent increase of anti-M1 antibodies. AP-MS analysis comparing the composition of the M1-plasma protein network from naïve and immunized mice showed significant enrichment of 292 IgG peptides associated with 56 IgG chains in the immune mice. Despite the significant increase of bound IgGs, the levels of interacting plasma proteins were not significantly reduced in the immune mice. The results indicate that the antigen-specific polyclonal IgG against the M1 protein primarily targets epitopes outside the other plasma protein binding interfaces. In conclusion, this study demonstrates that AP-MS is a promising strategy to determine the relationship between antigen-specific antibodies and host-pathogen interaction networks that could be used to define subdominant protein regions of relevance for vaccine development.


Assuntos
Antígenos de Bactérias , Imunoglobulina G , Ligação Proteica , Streptococcus pyogenes , Animais , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/metabolismo , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Camundongos , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunidade Adaptativa , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Anticorpos Antibacterianos/imunologia , Mapas de Interação de Proteínas , Espectrometria de Massas , Proteínas de Transporte/metabolismo , Proteínas de Transporte/imunologia , Feminino , Interações Hospedeiro-Patógeno/imunologia
2.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35217608

RESUMO

Therapeutic strategies directed at the tumor surfaceome (TS), including checkpoint inhibitor blocking antibodies, antibody drug conjugates (ADCs), and chimeric antigen receptor T (CAR-T) cells, provide a new armament to fight cancer. However, a remaining bottleneck is the lack of strategies to comprehensively interrogate patient tumors for potential TS targets. Here, we have developed a platform (tumor surfaceome mapping [TS-MAP]) integrated with a newly curated TS classifier (SURFME) that allows profiling of primary 3D cultures and intact patient glioma tumors with preserved tissue architecture. Moreover, TS-MAP specifically identifies proteins capable of endocytosis as tractable targets for ADCs and other modalities requiring toxic payload internalization. In high-grade gliomas that remain among the most aggressive forms of cancer, we show that cellular spatial organization (2D vs. 3D) fundamentally transforms the surfaceome and endocytome (e.g., integrins, proteoglycans, semaphorins, and cancer stem cell markers) with general implications for target screening approaches, as exemplified by an ADC targeting EGFR. The TS-MAP platform was further applied to profile the surfaceome and endocytome landscape in a cohort of freshly resected gliomas. We found a highly diverse TS repertoire between patient tumors, not directly associated with grade and histology, which highlights the need for individualized approaches. Our data provide additional layers of understanding fundamental to the future development of immunotherapy strategies, as well as procedures for proteomics-based target identification and selection. The TS-MAP platform should be widely applicable in efforts aiming at a better understanding of how to harness the TS for personalized immunotherapy.


Assuntos
Neoplasias Encefálicas/patologia , Endocitose , Glioma/patologia , Linhagem Celular Tumoral , Humanos , Proteínas de Neoplasias/metabolismo , Proteômica/métodos
3.
Anal Chem ; 96(22): 9060-9068, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38701337

RESUMO

An important element of antibody-guided vaccine design is the use of neutralizing or opsonic monoclonal antibodies to define protective epitopes in their native three-dimensional conformation. Here, we demonstrate a multimodal mass spectrometry-based strategy for in-depth characterization of antigen-antibody complexes to enable the identification of protective epitopes using the cytolytic exotoxin Streptolysin O (SLO) from Streptococcus pyogenes as a showcase. We first discovered a monoclonal antibody with an undisclosed sequence capable of neutralizing SLO-mediated cytolysis. The amino acid sequence of both the antibody light and the heavy chain was determined using mass-spectrometry-based de novo sequencing, followed by chemical cross-linking mass spectrometry to generate distance constraints between the antibody fragment antigen-binding region and SLO. Subsequent integrative computational modeling revealed a discontinuous epitope located in domain 3 of SLO that was experimentally validated by hydrogen-deuterium exchange mass spectrometry and reverse engineering of the targeted epitope. The results show that the antibody inhibits SLO-mediated cytolysis by binding to a discontinuous epitope in domain 3, likely preventing oligomerization and subsequent secondary structure transitions critical for pore-formation. The epitope is highly conserved across >98% of the characterized S. pyogenes isolates, making it an attractive target for antibody-based therapy and vaccine design against severe streptococcal infections.


Assuntos
Proteínas de Bactérias , Epitopos , Espectrometria de Massas , Streptococcus pyogenes , Estreptolisinas , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/química , Estreptolisinas/química , Estreptolisinas/imunologia , Estreptolisinas/metabolismo , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/química , Epitopos/imunologia , Epitopos/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/química , Sequência de Aminoácidos , Modelos Moleculares
4.
PLoS Comput Biol ; 19(1): e1010457, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36668672

RESUMO

Generating and analyzing overlapping peptides through multienzymatic digestion is an efficient procedure for de novo protein using from bottom-up mass spectrometry (MS). Despite improved instrumentation and software, de novo MS data analysis remains challenging. In recent years, deep learning models have represented a performance breakthrough. Incorporating that technology into de novo protein sequencing workflows require machine-learning models capable of handling highly diverse MS data. In this study, we analyzed the requirements for assembling such generalizable deep learning models by systemcally varying the composition and size of the training set. We assessed the generated models' performances using two test sets composed of peptides originating from the multienzyme digestion of samples from various species. The peptide recall values on the test sets showed that the deep learning models generated from a collection of highly N- and C-termini diverse peptides generalized 76% more over the termini-restricted ones. Moreover, expanding the training set's size by adding peptides from the multienzymatic digestion with five proteases of several species samples led to a 2-3 fold generalizability gain. Furthermore, we tested the applicability of these multienzyme deep learning (MEM) models by fully de novo sequencing the heavy and light monomeric chains of five commercial antibodies (mAbs). MEMs extracted over 10000 matching and overlapped peptides across six different proteases mAb samples, achieving a 100% sequence coverage for 8 of the ten polypeptide chains. We foretell that the MEMs' proven improvements to de novo analysis will positively impact several applications, such as analyzing samples of high complexity, unknown nature, or the peptidomics field.


Assuntos
Aprendizado Profundo , Proteômica , Proteômica/métodos , Espectrometria de Massas em Tandem/métodos , Peptídeos/química , Análise de Sequência de Proteína/métodos , Peptídeo Hidrolases , Anticorpos Monoclonais
5.
Mol Cell Proteomics ; 20: 100113, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34139362

RESUMO

Severe coronavirus disease 2019 (COVID-19) can result in pneumonia and acute respiratory failure. Accumulation of mucus in the airways is a hallmark of the disease and can result in hypoxemia. Here, we show that quantitative proteome analysis of the sputum from severe patients with COVID-19 reveal high levels of neutrophil extracellular trap (NET) components, which was confirmed by microscopy. Extracellular DNA from excessive NET formation can increase sputum viscosity and lead to acute respiratory distress syndrome. Recombinant human DNase (Pulmozyme; Roche) has been shown to be beneficial in reducing sputum viscosity and improve lung function. We treated five patients pwith COVID-19 resenting acute symptoms with clinically approved aerosolized Pulmozyme. No adverse reactions to the drug were seen, and improved oxygen saturation and recovery in all severely ill patients with COVID-19 was observed after therapy. Immunofluorescence and proteome analysis of sputum and blood plasma samples after treatment revealed a marked reduction of NETs and a set of statistically significant proteome changes that indicate reduction of hemorrhage, plasma leakage and inflammation in the airways, and reduced systemic inflammatory state in the blood plasma of patients. Taken together, the results indicate that NETs contribute to acute respiratory failure in COVID-19 and that degrading NETs may reduce dependency on external high-flow oxygen therapy in patients. Targeting NETs using recombinant human DNase may have significant therapeutic implications in COVID-19 disease and warrants further studies.


Assuntos
Tratamento Farmacológico da COVID-19 , Desoxirribonuclease I/farmacologia , Armadilhas Extracelulares/metabolismo , Proteoma/análise , Idoso , Proteínas Sanguíneas/análise , COVID-19/metabolismo , COVID-19/terapia , Feminino , Imunofluorescência , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/farmacologia , Índice de Gravidade de Doença , Escarro/efeitos dos fármacos , Escarro/metabolismo , Escarro/virologia , Síndrome de Resposta Inflamatória Sistêmica/tratamento farmacológico , Síndrome de Resposta Inflamatória Sistêmica/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/virologia
6.
Infect Immun ; 90(2): e0046221, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34898252

RESUMO

Sepsis is a life-threatening complication of infection that is characterized by a dysregulated inflammatory state and disturbed hemostasis. Platelets are the main regulators of hemostasis, and they also respond to inflammation. The human pathogen Streptococcus pyogenes can cause local infection that may progress to sepsis. There are more than 200 serotypes of S. pyogenes defined according to sequence variations in the M protein. The M1 serotype is among 10 serotypes that are predominant in invasive infection. M1 protein can be released from the surface and has previously been shown to generate platelet, neutrophil, and monocyte activation. The platelet-dependent proinflammatory effects of other serotypes of M protein associated with invasive infection (M3, M5, M28, M49, and M89) are now investigated using a combination of multiparameter flow cytometry, enzyme-linked immunosorbent assay (ELISA), aggregometry, and quantitative mass spectrometry. We demonstrate that only M1, M3, and M5 protein serotypes can bind fibrinogen in plasma and mediate fibrinogen- and IgG-dependent platelet activation and aggregation, release of granule proteins, upregulation of CD62P to the platelet surface, and complex formation with neutrophils and monocytes. Neutrophil and monocyte activation, determined as upregulation of surface CD11b, is also mediated by M1, M3, and M5 protein serotypes, while M28, M49, and M89 proteins failed to mediate activation of platelets or leukocytes. Collectively, our findings reveal novel aspects of the immunomodulatory role of fibrinogen acquisition and platelet activation during streptococcal infections.


Assuntos
Sepse , Infecções Estreptocócicas , Fibrinogênio/metabolismo , Humanos , Ativação Plaquetária , Sorogrupo , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo
7.
Bioinformatics ; 37(24): 4871-4872, 2021 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-34128979

RESUMO

SUMMARY: Protein-protein interactions (PPIs) are central in many biological processes but difficult to characterize, especially in complex, unfractionated samples. Chemical cross-linking combined with mass spectrometry (MS) and computational modeling is gaining recognition as a viable tool in protein interaction studies. Here, we introduce Cheetah-MS, a web server for predicting the PPIs in a complex mixture of samples. It combines the capability and sensitivity of MS to analyze complex samples with the power and resolution of protein-protein docking. It produces the quaternary structure of the PPI of interest by analyzing tandem MS/MS data (also called MS2). Combining MS analysis and modeling increases the sensitivity and, importantly, facilitates the interpretation of the results. AVAILABILITY AND IMPLEMENTATION: Cheetah-MS is freely available as a web server at https://www.txms.org.


Assuntos
Acinonyx , Animais , Acinonyx/metabolismo , Espectrometria de Massas em Tandem , Computadores , Proteínas/química , Simulação por Computador
8.
Am J Pathol ; 191(8): 1398-1411, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34111430

RESUMO

Bronchiolitis obliterans syndrome, a common form of chronic lung allograft dysfunction, is the major limitation to long-term survival after lung transplantation. The histologic correlate is progressive, fibrotic occlusion of small airways, obliterative bronchiolitis lesions, which ultimately lead to organ failure. The molecular composition of these lesions is unknown. In this sutdy, the protein composition of the lesions in explanted lungs from four end-stage bronchiolitis obliterans syndrome patients was analyzed using laser-capture microdissection and optimized sample preparation protocols for mass spectrometry. Immunohistochemistry and immunofluorescence were used to determine the spatial distribution of commonly identified proteins on the tissue level, and protein signatures for 14 obliterative bronchiolitis lesions were established. A set of 39 proteins, identified in >75% of lesions, included distinct structural proteins (collagen types IV and VI) and cellular components (actins, vimentin, and tryptase). Each respective lesion exhibited a unique composition of proteins (on average, n = 66 proteins), thereby mirroring the morphologic variation of the lesions. Antibody-based staining confirmed these mass spectrometry-based findings. The 14 analyzed obliterative bronchiolitis lesions showed variations in their protein content, but also common features. This study provides molecular and morphologic insights into the development of chronic rejection after lung transplantation. The protein patterns in the lesions were correlated to pathways of extracellular matrix organization, tissue development, and wound healing processes.


Assuntos
Bronquiolite Obliterante/metabolismo , Bronquiolite Obliterante/patologia , Pulmão/patologia , Transplantes/metabolismo , Transplantes/patologia , Remodelação das Vias Aéreas , Humanos , Microdissecção e Captura a Laser , Transplante de Pulmão , Proteoma
9.
PLoS Comput Biol ; 17(1): e1008169, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411763

RESUMO

Streptococcus pyogenes (Group A streptococcus; GAS) is an important human pathogen responsible for mild to severe, life-threatening infections. GAS expresses a wide range of virulence factors, including the M family proteins. The M proteins allow the bacteria to evade parts of the human immune defenses by triggering the formation of a dense coat of plasma proteins surrounding the bacteria, including IgGs. However, the molecular level details of the M1-IgG interaction have remained unclear. Here, we characterized the structure and dynamics of this interaction interface in human plasma on the surface of live bacteria using integrative structural biology, combining cross-linking mass spectrometry and molecular dynamics (MD) simulations. We show that the primary interaction is formed between the S-domain of M1 and the conserved IgG Fc-domain. In addition, we show evidence for a so far uncharacterized interaction between the A-domain and the IgG Fc-domain. Both these interactions mimic the protein G-IgG interface of group C and G streptococcus. These findings underline a conserved scavenging mechanism used by GAS surface proteins that block the IgG-receptor (FcγR) to inhibit phagocytic killing. We additionally show that we can capture Fab-bound IgGs in a complex background and identify XLs between the constant region of the Fab-domain and certain regions of the M1 protein engaged in the Fab-mediated binding. Our results elucidate the M1-IgG interaction network involved in inhibition of phagocytosis and reveal important M1 peptides that can be further investigated as future vaccine targets.


Assuntos
Antígenos de Bactérias , Proteínas da Membrana Bacteriana Externa , Proteínas de Transporte , Imunoglobulina G , Streptococcus pyogenes , Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Espectrometria de Massas , Simulação de Dinâmica Molecular , Fagocitose , Ligação Proteica , Streptococcus pyogenes/química , Streptococcus pyogenes/metabolismo , Fatores de Virulência/química , Fatores de Virulência/metabolismo
10.
J Proteome Res ; 20(5): 2983-3001, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33855848

RESUMO

Proteogenomic approaches have enabled the generat̲ion of novel information levels when compared to single omics studies although burdened by extensive experimental efforts. Here, we improved a data-independent acquisition mass spectrometry proteogenomic workflow to reveal distinct molecular features related to mammographic appearances in breast cancer. Our results reveal splicing processes detectable at the protein level and highlight quantitation and pathway complementarity between RNA and protein data. Furthermore, we confirm previously detected enrichments of molecular pathways associated with estrogen receptor-dependent activity and provide novel evidence of epithelial-to-mesenchymal activity in mammography-detected spiculated tumors. Several transcript-protein pairs displayed radically different abundances depending on the overall clinical properties of the tumor. These results demonstrate that there are differentially regulated protein networks in clinically relevant tumor subgroups, which in turn alter both cancer biology and the abundance of biomarker candidates and drug targets.


Assuntos
Neoplasias da Mama , Proteogenômica , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/genética , Feminino , Humanos , Mamografia , Fenótipo , Fluxo de Trabalho
11.
Anal Chem ; 93(8): 3929-3937, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33592145

RESUMO

We report a new design of an acoustophoretic trapping device with significantly increased capacity and throughput, compared to current commercial acoustic trapping systems. Acoustic trapping enables nanoparticle and extracellular vesicle (EV) enrichment without ultracentrifugation. Current commercial acoustic trapping technology uses an acoustic single-node resonance and typically operates at flow rates <50 µL/min, which limits the processing of the larger samples. Here, we use a larger capillary that supports an acoustic multinode resonance, which increased the seed particle capacity 40 times and throughput 25-40 times compared to single-node systems. The resulting increase in capacity and throughput was demonstrated by isolation of nanogram amounts of microRNA from acoustically trapped urinary EVs within 10 min. Additionally, the improved trapping performance enabled isolation of extracellular vesicles for downstream mass spectrometry analysis. This was demonstrated by the differential protein abundance profiling of urine samples (1-3 mL), derived from the non-trapped versus trapped urine samples.


Assuntos
Micropartículas Derivadas de Células , Vesículas Extracelulares , MicroRNAs , Acústica , Proteômica
12.
Diabet Med ; 38(11): e14658, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34309080

RESUMO

AIMS: Diabetic peripheral neuropathy (DPN) is a common and severe complication to type 2 diabetes. The pathogenesis of DPN is not fully known, but several pathways and gene polymorphisms contributing to DPN are described. DPN can be studied using nerve biopsies, but studies on the proteome of the nerve itself, and its surrounding tissue as a whole, are lacking. Studies on the posterior interosseous nerve (PIN) have proposed PIN a useful indicator of DPN. METHODS: A quantitative mass spectrometry-based proteomics analysis was made of peripheral nerves from age- and gender-matched living human male tissue donors; nine type 2 diabetes subjects, with decreased sural nerve action potentials indicating DPN, and six controls without type 2 diabetes, with normal electrophysiology results. RESULTS: A total of 2617 proteins were identified. Linear regression was used to discover which proteins were differentially expressed between type 2 diabetes and controls. Only soft signals were found. Therefore, clustering of the 500 most variable proteins was made to find clusters of similar proteins in type 2 diabetes subjects and healthy controls. CONCLUSIONS: This feasibility study shows, for the first time, that the use of quantitative mass spectrometry enables quantification of proteins from nerve biopsies from subjects with and without type 2 diabetes, which may aid in finding biomarkers of importance to DPN development.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Neuropatias Diabéticas/etiologia , Nervos Periféricos/fisiopatologia , Proteômica/métodos , Idoso , Diabetes Mellitus Tipo 2/epidemiologia , Neuropatias Diabéticas/epidemiologia , Feminino , Humanos , Incidência , Masculino , Suécia/epidemiologia
13.
J Immunol ; 202(2): 503-513, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30541884

RESUMO

Platelets circulate the bloodstream and principally maintain hemostasis. Disturbed hemostasis, a dysregulated inflammatory state, and a decreased platelet count are all hallmarks of severe invasive Streptococcus pyogenes infection, sepsis. We have previously demonstrated that the released M1 protein from S. pyogenes activates platelets, and this activation is dependent on the binding of M1 protein, fibrinogen, and M1-specific IgG to platelets in susceptible donors. In this study, we characterize the M1-associated protein interactions in human plasma and investigate the acquisition of proteins to the surface of activated platelets and the consequences for platelet immune function. Using quantitative mass spectrometry, M1 protein was determined to form a protein complex in plasma with statistically significant enrichment of fibrinogen, IgG3, and complement components, especially C1q. Using flow cytometry, these plasma proteins were also confirmed to be acquired to the platelet surface, resulting in complement activation on M1-activated human platelets. Furthermore, we demonstrated an increased phagocytosis of M1-activated platelets by monocytes, which was not observed with other physiological platelet agonists. This reveals a novel mechanism of complement activation during streptococcal sepsis, which contributes to the platelet consumption that occurs in sepsis.


Assuntos
Plaquetas/imunologia , Membrana Celular/metabolismo , Sepse/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/fisiologia , Anticorpos Antibacterianos/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/metabolismo , Células Cultivadas , Ativação do Complemento , Complemento C1q/metabolismo , Fibrinogênio/metabolismo , Citometria de Fluxo , Hemostasia , Humanos , Fagocitose , Ativação Plaquetária , Ligação Proteica
14.
Med Microbiol Immunol ; 209(3): 265-275, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32072248

RESUMO

A central challenge in infection medicine is to determine the structure and function of host-pathogen protein-protein interactions to understand how these interactions facilitate bacterial adhesion, dissemination and survival. In this review, we focus on proteomics, electron cryo-microscopy and structural modeling to showcase instances where affinity-purification (AP) and cross-linking (XL) mass spectrometry (MS) has advanced our understanding of host-pathogen interactions. We highlight cases where XL-MS in combination with structural modeling has provided insight into the quaternary structure of interspecies protein complexes. We further exemplify how electron cryo-tomography has been used to visualize bacterial-human interactions during attachment and infection. Lastly, we discuss how AP-MS, XL-MS and electron cryo-microscopy and -tomography together with structural modeling approaches can be used in future studies to broaden our knowledge regarding the function, dynamics and evolution of such interactions. This knowledge will be of relevance for future drug and vaccine development programs.


Assuntos
Interações entre Hospedeiro e Microrganismos , Modelos Moleculares , Mapeamento de Interação de Proteínas , Proteômica , Proteínas de Bactérias/química , Microscopia Crioeletrônica , Humanos , Espectrometria de Massas , Mapas de Interação de Proteínas , Estrutura Quaternária de Proteína
15.
Med Microbiol Immunol ; 209(3): 277-299, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31784893

RESUMO

The capacity of pathogenic microorganisms to adhere to host cells and avoid clearance by the host immune system is the initial and most decisive step leading to infections. Bacteria have developed different strategies to attach to diverse host surface structures. One important strategy is the adhesion to extracellular matrix (ECM) proteins (e.g., collagen, fibronectin, laminin) that are highly abundant in connective tissue and basement membranes. Gram-negative bacteria express variable outer membrane proteins (adhesins) to attach to the host and to initiate the process of infection. Understanding the underlying molecular mechanisms of bacterial adhesion is a prerequisite for targeting this interaction by "anti-ligands" to prevent colonization or infection of the host. Future development of such "anti-ligands" (specifically interfering with bacteria-host matrix interactions) might result in the development of a new class of anti-infective drugs for the therapy of infections caused by multidrug-resistant Gram-negative bacteria. This review summarizes our current knowledge about the manifold interactions of adhesins expressed by Gram-negative bacteria with ECM proteins and the use of this information for the generation of novel therapeutic antivirulence strategies.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana , Proteínas da Matriz Extracelular/fisiologia , Fibronectinas/fisiologia , Bactérias Gram-Negativas/fisiologia , Interações entre Hospedeiro e Microrganismos , Bactérias Gram-Negativas/patogenicidade , Humanos
16.
Mol Cell Proteomics ; 17(6): 1055-1066, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28389583

RESUMO

Cross-linking mass spectrometry (CLMS) provides distance constraints to study the structure of proteins, multiprotein complexes and protein-protein interactions which are critical for the understanding of protein function. CLMS is an attractive technology to bridge the gap between high-resolution structural biology techniques and proteomic-based interactome studies. However, as outlined in this review there are still several bottlenecks associated with CLMS which limit its application on a proteome-wide level. Specifically, there is an unmet need for comprehensive software that can reliably identify cross-linked peptides from large data sets. In this review we provide supporting information to reason that targeted proteomics of cross-links may provide the required sensitivity to reliably detect and quantify cross-linked peptides and that a reporter ion signature for cross-linked peptides may become a useful approach to increase confidence in the identification process of cross-linked peptides. In addition, the review summarizes the recent advances in CLMS workflows using the analysis of condensin complex in intact chromosomes as a model complex.


Assuntos
Espectrometria de Massas , Proteômica , Peptídeos , Mapeamento de Interação de Proteínas
17.
Mol Cell Proteomics ; 17(6): 1097-1111, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29511047

RESUMO

Infectious diseases are characterized by a complex interplay between host and pathogen, but how these interactions impact the host proteome is unclear. Here we applied a combined mass spectrometry-based proteomics strategy to investigate how the human proteome is transiently modified by the pathogen Streptococcus pyogenes, with a particular focus on bacterial cleavage of IgG in vivo In invasive diseases, S. pyogenes evokes a massive host response in blood, whereas superficial diseases are characterized by a local leakage of several blood plasma proteins at the site of infection including IgG. S. pyogenes produces IdeS, a protease cleaving IgG in the lower hinge region and we find highly effective IdeS-cleavage of IgG in samples from local IgG poor microenvironments. The results show that IdeS contributes to the adaptation of S. pyogenes to its normal ecological niches. Additionally, the work identifies novel clinical opportunities for in vivo pathogen detection.


Assuntos
Proteínas de Bactérias/farmacologia , Imunoglobulina G/metabolismo , Proteoma , Sepse/metabolismo , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes , Adolescente , Adulto , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Proteólise , Adulto Jovem
18.
BMC Bioinformatics ; 20(Suppl 4): 141, 2019 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-30999854

RESUMO

BACKGROUND: Bacterial surfaces are complex systems, constructed from membranes, peptidoglycan and, importantly, proteins. The proteins play crucial roles as critical regulators of how the bacterium interacts with and survive in its environment. A full catalog of the motifs in protein families and their relative conservation grade is a prerequisite to target the protein-protein interaction that bacterial surface protein makes to host proteins. RESULTS: In this paper, we propose a greedy approach to identify conserved motifs in large sequence families iteratively. Each iteration discovers a motif de novo and masks all occurrences of that motif. Remaining unmasked sequences are subjected to the next round of motif detection until no more significant motifs can be found. We demonstrate the utility of the method through the construction of a proteome-wide motif repository for Group A Streptococcus (GAS), a significant human pathogen. GAS produce numerous surface proteins that interact with over 100 human plasma proteins, helping the bacteria to evade the host immune response. We used the repository to find that proteins part of the bacterial surface has motif architectures that differ from intracellular proteins. CONCLUSIONS: We elucidate that the M protein, a coiled-coil homodimer that extends over 500 A from the cell wall, has a motif architecture that differs between various GAS strains. As the M protein is known to bind a variety of different plasma proteins, the results indicate that the different motif architectures are responsible for the quantitative differences of plasma proteins that various strains bind. The speed and applicability of the method enable its application to all major human pathogens.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Biologia Computacional/métodos , Proteoma/metabolismo , Algoritmos , Motivos de Aminoácidos , Sequência Conservada , Genoma Bacteriano , Streptococcus pyogenes/genética
19.
Int J Med Microbiol ; 309(7): 151325, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31257068

RESUMO

Aerococcus urinae is an emerging pathogen that causes urinary tract infections, bacteremia and infective endocarditis. The mechanisms through which A. urinae cause infection are largely unknown. The aims of this study were to describe the surface proteome of A. urinae and to analyse A. urinae genomes in search for genes encoding surface proteins. Two proteins, denoted Aerococcal surface protein (Asp) 1 and 2, were through the use of mass spectrometry based proteomics found to quantitatively dominate the aerococcal surface. The presence of these proteins on the surface was also shown using ELISA with serum from rabbits immunized with the recombinant Asp. These proteins had a signal sequence in the amino-terminal end and a cell wall-sorting region in the carboxy-terminal end, which contained an LPATG-motif, a hydrophobic domain and a positively charged tail. Twenty-three additional A. urinae genomes were sequenced using Illumina HiSeq technology. Six different variants of asp genes were found (denoted asp1-6). All isolates had either one or two of these asp-genes located in a conserved locus, designated Locus encoding Aerococcal Surface Proteins (LASP). The 25 genomes had in median 13 genes encoding LPXTG-proteins (range 6-24). For other Gram-positive bacteria, cell wall-anchored surface proteins with an LPXTG-motif play a key role for virulence. Thus, it will be of great interest to explore the function of the Asp proteins of A. urinae to establish a better understanding of the molecular mechanisms by which A. urinae cause disease.


Assuntos
Aerococcus/química , Proteínas de Bactérias/metabolismo , Parede Celular/química , Proteínas de Membrana/metabolismo , Aerococcus/genética , Aerococcus/metabolismo , Aerococcus/patogenicidade , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sequência de Bases , Parede Celular/metabolismo , Ensaio de Imunoadsorção Enzimática , Genoma Bacteriano/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Sinais Direcionadores de Proteínas , Proteoma , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Virulência/genética
20.
Cell Microbiol ; 20(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973822

RESUMO

Streptococcus pyogenes of the M1 serotype is commonly associated with invasive streptococcal infections and development of streptococcal toxic shock syndrome. The M1 protein is a powerful inducer of inflammatory responses for several human cell types, but the reason why M1 protein-related strains is over-represented in invasive streptococcal diseases is still not understood. This study was undertaken to investigate if soluble M1 protein can aggravate the severity of streptococcal skin infections in respect to inflammation, leucocyte recruitment, and tissue remodelling as seen in patients with cellulitis and necrotizing fasciitis. We found that HaCaT cells are able to recruit activated leucocytes when encountering M1 protein. Neither the bacterial protein nor activated leucocytes caused cell damage on HaCaT cells, instead HaCaT cells responded to the bacterial virulence factor by releasing several proteins protective against bacterial infection and leucocyte responses. However, although not cytotoxic, M1 protein completely abolished wound healing abilities of HaCaT cells. Taken together, our results demonstrate that M1 protein is a critical virulence factor that can augment streptococcal skin infection suggesting that the protein is an interesting target for drug development.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/imunologia , Celulite (Flegmão)/patologia , Fasciite Necrosante/patologia , Leucócitos/imunologia , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/patogenicidade , Linhagem Celular , Movimento Celular/fisiologia , Proliferação de Células , Celulite (Flegmão)/microbiologia , Fasciite Necrosante/microbiologia , Humanos , Queratinócitos , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/metabolismo , Fatores de Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA