Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell ; 186(23): 5041-5053.e19, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37865089

RESUMEN

To understand the molecular mechanisms of cellular pathways, contemporary workflows typically require multiple techniques to identify proteins, track their localization, and determine their structures in vitro. Here, we combined cellular cryoelectron tomography (cryo-ET) and AlphaFold2 modeling to address these questions and understand how mammalian sperm are built in situ. Our cellular cryo-ET and subtomogram averaging provided 6.0-Å reconstructions of axonemal microtubule structures. The well-resolved tertiary structures allowed us to unbiasedly match sperm-specific densities with 21,615 AlphaFold2-predicted protein models of the mouse proteome. We identified Tektin 5, CCDC105, and SPACA9 as novel microtubule-associated proteins. These proteins form an extensive interaction network crosslinking the lumen of axonemal doublet microtubules, suggesting their roles in modulating the mechanical properties of the filaments. Indeed, Tekt5 -/- sperm possess more deformed flagella with 180° bends. Together, our studies presented a cellular visual proteomics workflow and shed light on the in vivo functions of Tektin 5.


Asunto(s)
Proteoma , Espermatozoides , Animales , Masculino , Ratones , Axonema/química , Microscopía por Crioelectrón/métodos , Flagelos/metabolismo , Microtúbulos/metabolismo , Semen , Espermatozoides/química , Proteoma/análisis
2.
Cell ; 182(3): 685-712.e19, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32645325

RESUMEN

The causative agent of the coronavirus disease 2019 (COVID-19) pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected millions and killed hundreds of thousands of people worldwide, highlighting an urgent need to develop antiviral therapies. Here we present a quantitative mass spectrometry-based phosphoproteomics survey of SARS-CoV-2 infection in Vero E6 cells, revealing dramatic rewiring of phosphorylation on host and viral proteins. SARS-CoV-2 infection promoted casein kinase II (CK2) and p38 MAPK activation, production of diverse cytokines, and shutdown of mitotic kinases, resulting in cell cycle arrest. Infection also stimulated a marked induction of CK2-containing filopodial protrusions possessing budding viral particles. Eighty-seven drugs and compounds were identified by mapping global phosphorylation profiles to dysregulated kinases and pathways. We found pharmacologic inhibition of the p38, CK2, CDK, AXL, and PIKFYVE kinases to possess antiviral efficacy, representing potential COVID-19 therapies.


Asunto(s)
Betacoronavirus/metabolismo , Infecciones por Coronavirus/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Neumonía Viral/metabolismo , Proteómica/métodos , Células A549 , Enzima Convertidora de Angiotensina 2 , Animales , Antivirales/farmacología , COVID-19 , Células CACO-2 , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/metabolismo , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Pandemias , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Fosforilación , Neumonía Viral/virología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Vero , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Tirosina Quinasa del Receptor Axl
3.
Cell ; 169(4): 679-692.e14, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475896

RESUMEN

The nuclear RNA exosome is an essential multi-subunit complex that controls RNA homeostasis. Congenital mutations in RNA exosome genes are associated with neurodegenerative diseases. Little is known about the role of the RNA exosome in the cellular response to pathogens. Here, using NGS and human and mouse genetics, we show that influenza A virus (IAV) ribogenesis and growth are suppressed by impaired RNA exosome activity. Mechanistically, the nuclear RNA exosome coordinates the initial steps of viral transcription with RNAPII at host promoters. The viral polymerase complex co-opts the nuclear RNA exosome complex and cellular RNAs en route to 3' end degradation. Exosome deficiency uncouples chromatin targeting of the viral polymerase complex and the formation of cellular:viral RNA hybrids, which are essential RNA intermediates that license transcription of antisense genomic viral RNAs. Our results suggest that evolutionary arms races have shaped the cellular RNA quality control machinery.


Asunto(s)
Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Gripe Humana/virología , ARN Polimerasa II/metabolismo , Células A549 , Animales , Inmunoprecipitación de Cromatina , Exorribonucleasas/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Exosomas/metabolismo , Humanos , Espectrometría de Masas , Ratones , Mutación , Enfermedades Neurodegenerativas/virología , Proteínas de Unión al ARN/genética , Ribosomas/genética , Transcripción Genética
4.
Nat Rev Genet ; 23(6): 342-354, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35013567

RESUMEN

Understanding the effects of genetic variation is a fundamental problem in biology that requires methods to analyse both physical and functional consequences of sequence changes at systems-wide and mechanistic scales. To achieve a systems view, protein interaction networks map which proteins physically interact, while genetic interaction networks inform on the phenotypic consequences of perturbing these protein interactions. Until recently, understanding the molecular mechanisms that underlie these interactions often required biophysical methods to determine the structures of the proteins involved. The past decade has seen the emergence of new approaches based on coevolution, deep mutational scanning and genome-scale genetic or chemical-genetic interaction mapping that enable modelling of the structures of individual proteins or protein complexes. Here, we review the emerging use of large-scale genetic datasets and deep learning approaches to model protein structures and their interactions, and discuss the integration of structural data from different sources.


Asunto(s)
Mapas de Interacción de Proteínas , Proteínas , Epistasis Genética , Redes Reguladoras de Genes , Mutación , Mapeo de Interacción de Proteínas , Proteínas/genética , Proteínas/metabolismo
5.
Nat Rev Genet ; 21(6): 339-354, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32060427

RESUMEN

Ongoing social, political and ecological changes in the 21st century have placed more people at risk of life-threatening acute and chronic infections than ever before. The development of new diagnostic, prophylactic, therapeutic and curative strategies is critical to address this burden but is predicated on a detailed understanding of the immensely complex relationship between pathogens and their hosts. Traditional, reductionist approaches to investigate this dynamic often lack the scale and/or scope to faithfully model the dual and co-dependent nature of this relationship, limiting the success of translational efforts. With recent advances in large-scale, quantitative omics methods as well as in integrative analytical strategies, systems biology approaches for the study of infectious disease are quickly forming a new paradigm for how we understand and model host-pathogen relationships for translational applications. Here, we delineate a framework for a systems biology approach to infectious disease in three parts: discovery - the design, collection and analysis of omics data; representation - the iterative modelling, integration and visualization of complex data sets; and application - the interpretation and hypothesis-based inquiry towards translational outcomes.


Asunto(s)
Enfermedades Transmisibles/terapia , Interacciones Huésped-Patógeno/fisiología , Biología de Sistemas/métodos , Análisis de Datos , Humanos , Modelos Biológicos , Análisis de Sistemas
6.
Nature ; 583(7816): 459-468, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32353859

RESUMEN

A newly described coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is the causative agent of coronavirus disease 2019 (COVID-19), has infected over 2.3 million people, led to the death of more than 160,000 individuals and caused worldwide social and economic disruption1,2. There are no antiviral drugs with proven clinical efficacy for the treatment of COVID-19, nor are there any vaccines that prevent infection with SARS-CoV-2, and efforts to develop drugs and vaccines are hampered by the limited knowledge of the molecular details of how SARS-CoV-2 infects cells. Here we cloned, tagged and expressed 26 of the 29 SARS-CoV-2 proteins in human cells and identified the human proteins that physically associated with each of the SARS-CoV-2 proteins using affinity-purification mass spectrometry, identifying 332 high-confidence protein-protein interactions between SARS-CoV-2 and human proteins. Among these, we identify 66 druggable human proteins or host factors targeted by 69 compounds (of which, 29 drugs are approved by the US Food and Drug Administration, 12 are in clinical trials and 28 are preclinical compounds). We screened a subset of these in multiple viral assays and found two sets of pharmacological agents that displayed antiviral activity: inhibitors of mRNA translation and predicted regulators of the sigma-1 and sigma-2 receptors. Further studies of these host-factor-targeting agents, including their combination with drugs that directly target viral enzymes, could lead to a therapeutic regimen to treat COVID-19.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/metabolismo , Reposicionamiento de Medicamentos , Terapia Molecular Dirigida , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/metabolismo , Mapas de Interacción de Proteínas , Proteínas Virales/metabolismo , Animales , Antivirales/clasificación , Antivirales/farmacología , Betacoronavirus/genética , Betacoronavirus/metabolismo , Betacoronavirus/patogenicidad , COVID-19 , Chlorocebus aethiops , Clonación Molecular , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Evaluación Preclínica de Medicamentos , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Inmunidad Innata , Espectrometría de Masas , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/virología , Unión Proteica , Biosíntesis de Proteínas/efectos de los fármacos , Dominios Proteicos , Mapeo de Interacción de Proteínas , Receptores sigma/metabolismo , SARS-CoV-2 , Proteínas Ligasas SKP Cullina F-box/metabolismo , Células Vero , Proteínas Virales/genética , Tratamiento Farmacológico de COVID-19
7.
Mol Cell Proteomics ; 23(5): 100755, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38548018

RESUMEN

Human APOBEC3 enzymes are a family of single-stranded (ss)DNA and RNA cytidine deaminases that act as part of the intrinsic immunity against viruses and retroelements. These enzymes deaminate cytosine to form uracil which can functionally inactivate or cause degradation of viral or retroelement genomes. In addition, APOBEC3s have deamination-independent antiviral activity through protein and nucleic acid interactions. If expression levels are misregulated, some APOBEC3 enzymes can access the human genome leading to deamination and mutagenesis, contributing to cancer initiation and evolution. While APOBEC3 enzymes are known to interact with large ribonucleoprotein complexes, the function and RNA dependence are not entirely understood. To further understand their cellular roles, we determined by affinity purification mass spectrometry (AP-MS) the protein interaction network for the human APOBEC3 enzymes and mapped a diverse set of protein-protein and protein-RNA mediated interactions. Our analysis identified novel RNA-mediated interactions between APOBEC3C, APOBEC3H Haplotype I and II, and APOBEC3G with spliceosome proteins, and APOBEC3G and APOBEC3H Haplotype I with proteins involved in tRNA methylation and ncRNA export from the nucleus. In addition, we identified RNA-independent protein-protein interactions with APOBEC3B, APOBEC3D, and APOBEC3F and the prefoldin family of protein-folding chaperones. Interaction between prefoldin 5 (PFD5) and APOBEC3B disrupted the ability of PFD5 to induce degradation of the oncogene cMyc, implicating the APOBEC3B protein interaction network in cancer. Altogether, the results uncover novel functions and interactions of the APOBEC3 family and suggest they may have fundamental roles in cellular RNA biology, their protein-protein interactions are not redundant, and there are protein-protein interactions with tumor suppressors, suggesting a role in cancer biology. Data are available via ProteomeXchange with the identifier PXD044275.


Asunto(s)
Citidina Desaminasa , Mapas de Interacción de Proteínas , Humanos , Citidina Desaminasa/metabolismo , Citidina Desaminasa/genética , Desaminación , Desaminasas APOBEC/metabolismo , Aminohidrolasas/metabolismo , Aminohidrolasas/genética , Células HEK293 , Citosina Desaminasa/metabolismo , Desaminasa APOBEC-3G/metabolismo , Desaminasa APOBEC-3G/genética , Empalmosomas/metabolismo , Unión Proteica , Espectrometría de Masas , ARN/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Antígenos de Histocompatibilidad Menor/genética
8.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34349018

RESUMEN

Defining protein-protein interactions (PPIs) in their native environment is crucial to understanding protein structure and function. Cross-linking-mass spectrometry (XL-MS) has proven effective in capturing PPIs in living cells; however, the proteome coverage remains limited. Here, we have developed a robust in vivo XL-MS platform to facilitate in-depth PPI mapping by integrating a multifunctional MS-cleavable cross-linker with sample preparation strategies and high-resolution MS. The advancement of click chemistry-based enrichment significantly enhanced the detection of cross-linked peptides for proteome-wide analyses. This platform enabled the identification of 13,904 unique lysine-lysine linkages from in vivo cross-linked HEK 293 cells, permitting construction of the largest in vivo PPI network to date, comprising 6,439 interactions among 2,484 proteins. These results allowed us to generate a highly detailed yet panoramic portrait of human interactomes associated with diverse cellular pathways. The strategy presented here signifies a technological advancement for in vivo PPI mapping at the systems level and can be generalized for charting protein interaction landscapes in any organisms.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Espectrometría de Masas/métodos , Mapeo de Interacción de Proteínas/métodos , Chaperoninas/análisis , Chaperoninas/química , Chaperoninas/metabolismo , Química Clic/métodos , Células HEK293 , Histonas/metabolismo , Humanos , Lisina/química , Complejos Multiproteicos/química , Péptidos/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica/métodos , Reproducibilidad de los Resultados , Ubiquitina/metabolismo
9.
Mol Cell Proteomics ; 20: 100132, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34389466

RESUMEN

Structural analysis of host-pathogen protein complexes remains challenging, largely due to their structural heterogeneity. Here, we describe a pipeline for the structural characterization of these complexes using integrative structure modeling based on chemical cross-links and residue-protein contacts inferred from mutagenesis studies. We used this approach on the HIV-1 Vif protein bound to restriction factor APOBEC3G (A3G), the Cullin-5 E3 ring ligase (CRL5), and the cellular transcription factor Core Binding Factor Beta (CBFß) to determine the structure of the (A3G-Vif-CRL5-CBFß) complex. Using the MS-cleavable DSSO cross-linker to obtain a set of 132 cross-links within this reconstituted complex along with the atomic structures of the subunits and mutagenesis data, we computed an integrative structure model of the heptameric A3G-Vif-CRL5-CBFß complex. The structure, which was validated using a series of tests, reveals that A3G is bound to Vif mostly through its N-terminal domain. Moreover, the model ensemble quantifies the dynamic heterogeneity of the A3G C-terminal domain and Cul5 positions. Finally, the model was used to rationalize previous structural, mutagenesis and functional data not used for modeling, including information related to the A3G-bound and unbound structures as well as mapping functional mutations to the A3G-Vif interface. The experimental and computational approach described here is generally applicable to other challenging host-pathogen protein complexes.


Asunto(s)
Desaminasa APOBEC-3G/química , Subunidad beta del Factor de Unión al Sitio Principal/química , Proteínas Cullin/química , Ubiquitina-Proteína Ligasas/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química , Espectrometría de Masas , Modelos Moleculares
10.
J Proteome Res ; 20(2): 1133-1152, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33464917

RESUMEN

The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), was declared a pandemic infection in March 2020. As of December 2020, two COVID-19 vaccines have been authorized for emergency use by the U.S. Food and Drug Administration, but there are no effective drugs to treat COVID-19, and pandemic mitigation efforts like physical distancing have had acute social and economic consequences. In this perspective, we discuss how the proteomic research community can leverage technologies and expertise to address the pandemic by investigating four key areas of study in SARS-CoV-2 biology. Specifically, we discuss how (1) mass spectrometry-based structural techniques can overcome limitations and complement traditional structural approaches to inform the dynamic structure of SARS-CoV-2 proteins, complexes, and virions; (2) virus-host protein-protein interaction mapping can identify the cellular machinery required for SARS-CoV-2 replication; (3) global protein abundance and post-translational modification profiling can characterize signaling pathways that are rewired during infection; and (4) proteomic technologies can aid in biomarker identification, diagnostics, and drug development in order to monitor COVID-19 pathology and investigate treatment strategies. Systems-level high-throughput capabilities of proteomic technologies can yield important insights into SARS-CoV-2 biology that are urgently needed during the pandemic, and more broadly, can inform coronavirus virology and host biology.


Asunto(s)
COVID-19/prevención & control , Proteoma/metabolismo , Proteómica/métodos , SARS-CoV-2/metabolismo , COVID-19/epidemiología , COVID-19/virología , Interacciones Huésped-Patógeno , Humanos , Espectrometría de Masas/métodos , Pandemias , Mapas de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , SARS-CoV-2/fisiología , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA