Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2659: 171-182, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37249893

RESUMO

Quantitative proteomics is a powerful method for distinguishing protein abundance changes in a biological system across conditions. In addition to recent advances in computational power and bioinformatics methods, improvements to sensitivity and resolution of mass spectrometry (MS) instrumentation provide an innovative approach for studying host-pathogen interaction dynamics and posttranslational modifications. In this protocol, we provide a workflow for state-of-the-art MS-based proteomics to assess changes in phosphorylated protein abundance upon interaction between the worldwide cereal crop, Triticum aestivum (wheat), and the global cereal crop fungal pathogen, Fusarium graminearum, during infection. This protocol mimics a time course of infection of T. aestivum by F. graminearum in the greenhouse, and the harvested samples undergo Fe-NTA phosphoenrichment combined with label-free quantification (LFQ) for detection by liquid-chromatography (LC)-coupled with tandem MS/MS. Our approach provides an in-depth view of changes in phosphorylation from both the host and pathogen perspectives in a single experiment across infection time points and different host cultivars.


Assuntos
Fusarium , Triticum , Triticum/microbiologia , Espectrometria de Massas em Tandem , Doenças das Plantas/microbiologia , Proteômica , Fusarium/metabolismo , Proteoma/metabolismo
2.
mBio ; 13(4): e0168722, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35862772

RESUMO

The interactions between a host and microbe drive the health and disease status of the host. Of importance is the cause of dysbiosis in the presence of a pathogen, and critically, the relationship between the host and pathogen may evolve over time through response and adaptation. For immunocompromised individuals, dual infections are prevalent and contribute to disease severity and treatment options. Here, we explore the global reprogramming of host cells in response to immediate and established microbial infections with the human fungal pathogen Cryptococcus neoformans and the nosocomial bacterial pathogen Klebsiella pneumoniae. Using quantitative proteomics, we uncovered cross-kingdom protein-level changes associated with initial fungal infection, followed by a remarkable adaptation of the host and pathogen to a dormant state. This stabilization is disrupted over time upon bacterial infection, with the production of virulence-associated bacterial proteins and severely altered host response. We support our findings with the profiling of two major virulence determinants in C. neoformans, catalase and melanin, which demonstrate an interconnected regulation in response to both host defense and bacterial invasion. Overall, we report novel fungal and bacterial modulation of the host, including adaptation and stabilization, suggesting an opportunity to effectively treat dual infections by selectively targeting proteins critical to the host's infection stage. IMPORTANCE The relationship between the human microbiota and infectious disease outcome is a rapidly expanding area of study. Understanding how the host responds to changes in its symbiotic relationship with microbes provides new insight into how disruption can promote disease. In this study, we investigated the evolving relationship between innate immune cells of the host during immediate and established infections with fungal and bacterial pathogens, commonly observed within the lungs of immunocompromised individuals. We observed critical reprogramming of each biological system over time and in response to the changing environment, which influences microbial virulence. The goal of this important work is to improve our fundamental understanding of pathogenesis, as well as the regulatory relationships between hosts and microbes that drive disease outcome. We envision defining improved therapeutic treatment options for the host dependent on disease state to reduce the global impact and burden of infectious diseases, especially in the face of ever-increasing rates of antimicrobial resistance.


Assuntos
Infecção Hospitalar , Criptococose , Cryptococcus neoformans , Criptococose/microbiologia , Humanos , Macrófagos/microbiologia , Virulência
3.
Methods Mol Biol ; 2456: 141-151, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35612740

RESUMO

Phosphorylation is a key post-translational modification central to the biological behavior of proteins. This reversible modification specifically regulates cell signaling mechanisms to control survival and growth. Moreover, microbial pathogens, including both fungi and bacteria, rely on this modification to coordinate protein production and functioning during infection and dissemination within a host. Understanding phosphorylation and its involvement with effector proteins and complex networks are now possible with the recent technological advancements of mass spectrometry. Herein, we describe a phosphopeptide enrichment strategy optimized for the invasive mycosis-causing fungal pathogen Cryptococcus neoformans. Our protocol details proper sample preparation for efficient lysis and protein extraction with minimal phosphorylation losses followed by outlined steps for enrichment, instrumentation handling, and data analysis to permit deep profiling of the global phosphoproteome. The high-throughput versatility of bottom-up proteomics combined with our sample preparation approach facilitates opportunities for in-depth phosphorylation mapping and novel biological discoveries.


Assuntos
Criptococose , Cryptococcus neoformans , Criptococose/microbiologia , Fosfopeptídeos/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteoma/metabolismo , Proteômica/métodos
4.
J Fungi (Basel) ; 7(2)2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33572187

RESUMO

Post-translational modifications (PTMs) change the structure and function of proteins and regulate a diverse array of biological processes. Fungal pathogens rely on PTMs to modulate protein production and activity during infection, manipulate the host response, and ultimately, promote fungal survival. Given the high mortality rates of fungal infections on a global scale, along with the emergence of antifungal-resistant species, identifying new treatment options is critical. In this review, we focus on the role of PTMs (e.g., phosphorylation, acetylation, ubiquitination, glycosylation, and methylation) among the highly prevalent and medically relevant fungal pathogens, Candida spp., Aspergillus spp., and Cryptococcus spp. We explore the role of PTMs in fungal stress response and host adaptation, the use of PTMs to manipulate host cells and the immune system upon fungal invasion, and the importance of PTMs in conferring antifungal resistance. We also provide a critical view on the current knowledgebase, pose questions key to our understanding of the intricate roles of PTMs within fungal pathogens, and provide research opportunities to uncover new therapeutic strategies.

5.
J Vis Exp ; (164)2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33165315

RESUMO

The technological achievements of mass spectrometry (MS)-based quantitative proteomics opens many undiscovered avenues for analyzing an organism's global proteome under varying conditions. This powerful strategy applied to the interactions of microbial pathogens with the desired host comprehensively characterizes both perspectives towards infection. Herein, the workflow describes label-free quantification (LFQ) of the infectome of Cryptococcus neoformans, a fungal facultative intracellular pathogen that is the causative agent of the deadly disease cryptococcosis, in the presence of immortalized macrophage cells. The protocol details the proper protein preparation techniques for both pathogen and mammalian cells within a single experiment, resulting in appropriate peptide submission for liquid-chromatography (LC)-MS/MS analysis. The high throughput generic nature of LFQ allows a wide dynamic range of protein identification and quantification, as well as transferability to any host-pathogen infection setting, maintaining extreme sensitivity. The method is optimized to catalogue extensive, unbiased protein abundance profiles of a pathogen within infection-mimicking conditions. Specifically, the method demonstrated here provides essential information on C. neoformans pathogenesis, such as protein production necessary for virulence and identifies critical host proteins responding to microbial invasion.


Assuntos
Interações Hospedeiro-Patógeno , Proteômica/métodos , Fluxo de Trabalho , Animais , Linhagem Celular , Cryptococcus neoformans/fisiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Espectrometria de Massas em Tandem
6.
Curr Protoc Microbiol ; 55(1): e94, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31797572

RESUMO

Cryptococcus neoformans is an opportunistic human fungal pathogen commonly associated with infection in immunocompromised individuals (e.g., patients with HIV/AIDS). Important virulence determinants include the production of a polysaccharide capsule, melanin, and extracellular enzymes, as well as the ability to grow at 37°C. C. neoformans controls a plethora of host defense and evasion mechanisms to survive during infection and to proliferate within the host, causing meningoencephalitis and death. Traditionally, characterization of C. neoformans under different environmental conditions and stresses has relied on genetic and phenotypic analyses, as well as biochemical assays. However, advances in mass spectrometry instrumentation, sample preparation protocols, and bioinformatic tools and databases promote comprehensive profiling of fungal cellular processes, secretion or protein release into the extracellular environment, and vesicle contents. Moreover, proteomics provides insight into regulatory mechanisms influencing signal transduction cascades and protein complexes or networks through profiling of post-translational modifications and protein-protein interactions. Given the medical impact of C. neoformans infections and the recent emergence of antifungal-resistant strains, defining proteins produced in response to unique environments provides an opportunity to uncover antivirulence strategies and alternative therapeutic options to combat infection. Here, we describe culturing and sample preparation of C. neoformans and outline protocols for comprehensively profiling changes in protein abundance within the cellular proteome and secretome. © 2019 by John Wiley & Sons, Inc. Basic Protocol 1: Growth and sample preparation of Cryptococcus neoformans Basic Protocol 2: Protein extraction from supernatant Basic Protocol 3: Protein extraction from cell pellet Basic Protocol 4: Proteomic profiling and bioinformatics.


Assuntos
Cryptococcus neoformans/química , Proteínas Fúngicas/análise , Proteoma/análise , Proteômica/métodos , Biologia Computacional/métodos , Cryptococcus neoformans/crescimento & desenvolvimento , Espectrometria de Massas/métodos , Técnicas Microbiológicas/métodos , Manejo de Espécimes/métodos
7.
J Fungi (Basel) ; 5(2)2019 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-31212923

RESUMO

The prevalence of fungal diseases is increasing on a global scale, ranging from acute to systemic infections caused by commensal or pathogenic microorganisms, often associated with the immune status of the host. Morbidity and mortality rates remain high and our ability to treat fungal infections is challenged by a limited arsenal of antifungal agents and the emergence of drug resistant pathogens. There is a high demand for new approaches to elucidate the fungal mechanisms of pathogenesis and the interplay between host and pathogen to discover novel treatment options. Moreover, the need for improved drug efficacy and reduced host toxicity requires the identification and characterization of antifungal biological targets and molecular mechanisms of action. Mass spectrometry (MS)-based proteomics is a rapidly advancing field capable of addressing these priorities by providing comprehensive information on the dynamics of cellular processes, modifications, and interactions. In this Review, we focus on applications of MS-based proteomics in a diverse array of fungal pathogens and host systems to define and distinguish the molecular details of fungal pathogenesis and host-fungal interactions. We also explore the emerging role of MS-based proteomics in the discovery and development of novel antifungal therapies and provide insight into the future of MS-based proteomics in fungal biology.

8.
PLoS Biol ; 17(6): e3000334, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31206517

RESUMO

Escherichia coli represents a classical intestinal gram-negative commensal. Despite this commensalism, different E. coli strains can mediate disparate immunogenic properties in a given host. Symbiotic E. coli strains such as E. coli Nissle 1917 (EcN) are attributed beneficial properties, e.g., promotion of intestinal homeostasis. Therefore, we aimed to identify molecular features derived from symbiotic bacteria that might help to develop innovative therapeutic alternatives for the treatment of intestinal immune disorders. This study was performed using the dextran sodium sulphate (DSS)-induced colitis mouse model, which is routinely used to evaluate potential therapeutics for the treatment of Inflammatory Bowel Diseases (IBDs). We focused on the analysis of flagellin structures of different E. coli strains. EcN flagellin was found to harbor a substantially longer hypervariable region (HVR) compared to other commensal E. coli strains, and this longer HVR mediated symbiotic properties through stronger activation of Toll-like receptor (TLR)5, thereby resulting in interleukin (IL)-22-mediated protection of mice against DSS-induced colitis. Furthermore, using bone-marrow-chimeric mice (BMCM), CD11c+ cells of the colonic lamina propria (LP) were identified as the main mediators of these flagellin-induced symbiotic effects. We propose flagellin from symbiotic E. coli strains as a potential therapeutic to restore intestinal immune homeostasis, e.g., for the treatment of IBD patients.


Assuntos
Escherichia coli/metabolismo , Flagelina/genética , Simbiose/genética , Animais , Colite/induzido quimicamente , Colite/imunologia , Modelos Animais de Doenças , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Feminino , Flagelina/metabolismo , Mucosa Intestinal , Intestinos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Simbiose/fisiologia , Receptor 5 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...