Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Mol Syst Biol ; 20(8): 952-971, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38951684

RESUMEN

Proximity labeling (PL) via biotinylation coupled with mass spectrometry (MS) captures spatial proteomes in cells. Large-scale processing requires a workflow minimizing hands-on time and enhancing quantitative reproducibility. We introduced a scalable PL pipeline integrating automated enrichment of biotinylated proteins in a 96-well plate format. Combining this with optimized quantitative MS based on data-independent acquisition (DIA), we increased sample throughput and improved protein identification and quantification reproducibility. We applied this pipeline to delineate subcellular proteomes across various compartments. Using the 5HT2A serotonin receptor as a model, we studied temporal changes of proximal interaction networks induced by receptor activation. In addition, we modified the pipeline for reduced sample input to accommodate CRISPR-based gene knockout, assessing dynamics of the 5HT2A network in response to perturbation of selected interactors. This PL approach is universally applicable to PL proteomics using biotinylation-based PL enzymes, enhancing throughput and reproducibility of standard protocols.


Asunto(s)
Biotinilación , Proteoma , Proteómica , Proteómica/métodos , Reproducibilidad de los Resultados , Humanos , Proteoma/metabolismo , Espectrometría de Masas/métodos , Células HEK293
2.
ArXiv ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38800652

RESUMEN

Proteins congregate into complexes to perform fundamental cellular functions. Phenotypic outcomes, in health and disease, are often mechanistically driven by the remodeling of protein complexes by protein-coding mutations or cellular signaling changes in response to molecular cues. Here, we present an affinity purification-mass spectrometry (APMS) proteomics protocol to quantify and visualize global changes in protein-protein interaction (PPI) networks between pairwise conditions. We describe steps for expressing affinity-tagged "bait" proteins in mammalian cells, identifying purified protein complexes, quantifying differential PPIs, and visualizing differential PPI networks. Specifically, this protocol details steps for designing affinity-tagged "bait" gene constructs, transfection, affinity purification, mass spectrometry sample preparation, data acquisition, database search, data quality control, PPI confidence scoring, cross-run normalization, statistical data analysis, and differential PPI visualization. Our protocol discusses caveats and limitations with applicability across cell types and biological areas. For complete details on the use and execution of this protocol, please refer to Bouhaddou et al. 20231.

3.
Artículo en Inglés | MEDLINE | ID: mdl-38758153

RESUMEN

Context: Gastric cancer (GC) is a common and life-threatening gastrointestinal malignancy. Although mucin 3A (MUC3A) is an essential oncogenic factor in several cancers, limited information is available on its expression in GC tissues and its impact on prognosis. Objective: The study aimed to characterize MUC3A in GC and to explore its potential involvement in regulating GC cells' behavior through the mammalian target of rapamycin (mTOR) signaling pathway. Design: The research team conducted a retrospective genetic analysis. Setting: The study took place as Huzhou Central Hospital, an Affiliated Central Hospital of Huzhou University in Huzhou, Zhejiang, China. Participants: Participants were 47 patients with GC who had received treatment at the department of general surgery at the hospital and who gave consent for the use of their tissue samples for the genetic analysis. Outcome Measures: The research team: (1) performed a differential analysis of MUC3A using GC and normal tissue samples purchased from the American Type Culture Collection; (2) investigated the exposure of cancer tissues to MUC3A and its effects in the tumor, node, metastasis (TNM) stages of GC, using the real-time quantitative polymerase chain reaction (rt-qPCR) method; (3) performed clone formation and conducted transwell assays by knocking down or overexpressing MUC3A to analyze the effects on the behavior of GC cells; and (4) assessed the content of related marker proteins and the phosphoinositide 3-kinase (PI3K)/ protein kinase B (Akt)/ mammalian target of rapamycin (mTOR) pathway proteins, using a Western blot analysis. Results: A high level of MUC3A existed in GC tissues, and it was associated with TNM staging. Silencing of the MUC3A inhibited GC-cell migration and proliferation, and MUC3A overexpression had the opposite effect. The addition of agonist M05856 restored the inhibitory effect of silencing MUC3A on GC cell proliferation and migration, suggesting that MUC3A regulates GC cells' behavior through the PI3K/Akt/mTOR pathway. Conclusions: MUC3A plays an oncogenic role in GC and may regulate GC cell behavior through the PI3K/Akt/mTOR pathway.

4.
Nat Chem Biol ; 20(9): 1133-1143, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38528119

RESUMEN

The µ-opioid receptor (µOR) represents an important target of therapeutic and abused drugs. So far, most understanding of µOR activity has focused on a subset of known signal transducers and regulatory molecules. Yet µOR signaling is coordinated by additional proteins in the interaction network of the activated receptor, which have largely remained invisible given the lack of technologies to interrogate these networks systematically. Here we describe a proteomics and computational approach to map the proximal proteome of the activated µOR and to extract subcellular location, trafficking and functional partners of G-protein-coupled receptor (GPCR) activity. We demonstrate that distinct opioid agonists exert differences in the µOR proximal proteome mediated by endocytosis and endosomal sorting. Moreover, we identify two new µOR network components, EYA4 and KCTD12, which are recruited on the basis of receptor-triggered G-protein activation and might form a previously unrecognized buffering system for G-protein activity broadly modulating cellular GPCR signaling.


Asunto(s)
Proteoma , Proteómica , Receptores Opioides mu , Humanos , Endocitosis , Células HEK293 , Proteoma/metabolismo , Proteómica/métodos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Opioides mu/metabolismo , Receptores Opioides mu/agonistas , Transducción de Señal
5.
bioRxiv ; 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38076945

RESUMEN

Translating high-confidence (hc) autism spectrum disorder (ASD) genes into viable treatment targets remains elusive. We constructed a foundational protein-protein interaction (PPI) network in HEK293T cells involving 100 hcASD risk genes, revealing over 1,800 PPIs (87% novel). Interactors, expressed in the human brain and enriched for ASD but not schizophrenia genetic risk, converged on protein complexes involved in neurogenesis, tubulin biology, transcriptional regulation, and chromatin modification. A PPI map of 54 patient-derived missense variants identified differential physical interactions, and we leveraged AlphaFold-Multimer predictions to prioritize direct PPIs and specific variants for interrogation in Xenopus tropicalis and human forebrain organoids. A mutation in the transcription factor FOXP1 led to reconfiguration of DNA binding sites and altered development of deep cortical layer neurons in forebrain organoids. This work offers new insights into molecular mechanisms underlying ASD and describes a powerful platform to develop and test therapeutic strategies for many genetically-defined conditions.

6.
Cell ; 186(21): 4597-4614.e26, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37738970

RESUMEN

SARS-CoV-2 variants of concern (VOCs) emerged during the COVID-19 pandemic. Here, we used unbiased systems approaches to study the host-selective forces driving VOC evolution. We discovered that VOCs evolved convergent strategies to remodel the host by modulating viral RNA and protein levels, altering viral and host protein phosphorylation, and rewiring virus-host protein-protein interactions. Integrative computational analyses revealed that although Alpha, Beta, Gamma, and Delta ultimately converged to suppress interferon-stimulated genes (ISGs), Omicron BA.1 did not. ISG suppression correlated with the expression of viral innate immune antagonist proteins, including Orf6, N, and Orf9b, which we mapped to specific mutations. Later Omicron subvariants BA.4 and BA.5 more potently suppressed innate immunity than early subvariant BA.1, which correlated with Orf6 levels, although muted in BA.4 by a mutation that disrupts the Orf6-nuclear pore interaction. Our findings suggest that SARS-CoV-2 convergent evolution overcame human adaptive and innate immune barriers, laying the groundwork to tackle future pandemics.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/virología , Inmunidad Innata/genética , Pandemias , SARS-CoV-2/genética
7.
bioRxiv ; 2023 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-37090610

RESUMEN

Proximity labeling (PL) coupled with mass spectrometry has emerged as a powerful technique to map proximal protein interactions in living cells. Large-scale sample processing for proximity proteomics necessitates a high-throughput workflow to reduce hands-on time and increase quantitative reproducibility. To address this issue, we developed a scalable and automated PL pipeline, including generation and characterization of monoclonal cell lines, automated enrichment of biotinylated proteins in a 96-well format, and optimization of the quantitative mass spectrometry (MS) acquisition method. Combined with data-independent acquisition (DIA) MS, our pipeline outperforms manual enrichment and data-dependent acquisition (DDA) MS regarding reproducibility of protein identification and quantification. We apply the pipeline to map subcellular proteomes for endosomes, late endosomes/lysosomes, the Golgi apparatus, and the plasma membrane. Moreover, using serotonin receptor (5HT2A) as a model, we investigated agonist-induced dynamics in protein-protein interactions. Importantly, the approach presented here is universally applicable for PL proteomics using all biotinylation-based PL enzymes, increasing both throughput and reproducibility of standard protocols.

8.
Antimicrob Agents Chemother ; 67(5): e0170422, 2023 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-37052498

RESUMEN

Clostridioides difficile infection (CDI) causes severe diarrhea and colitis, leading to significant morbidity, mortality, and high medical costs worldwide. Oral vancomycin, a first-line treatment for CDI, is associated with a high risk of recurrence, necessitating novel therapies for primary and recurrent CDI. A novel small-molecule compound, CDBN-YGXZ, was synthesized by modifying the benzene ring of nitazoxanide with lauric acid. The mechanism of action of CDBN-YGXZ was validated using a pyruvate:ferredoxin/flavodoxin oxidoreductase (PFOR) inhibition assay. The efficacy of CDBN-YGXZ was evaluated using the MIC test and CDI infection model in mice and hamsters. Furthermore, metagenomics was used to reveal the underlying reasons for the effective reduction or prevention of CDI after CDBN-YGXZ treatment. The inhibitory activity against PFOR induced by CDBN-YGXZ. MIC tests showed that the in vitro activity of CDBN-YGXZ against C. difficile ranging from 0.1 to 1.5 µg/mL. In the mouse and hamster CDI models, CDBN-YGXZ provided protection during both treatment and relapse, while vancomycin treatment resulted in severe relapse and significant clinical scores. Compared with global effects on the indigenous gut microbiota induced by vancomycin, CDBN-YGXZ treatment had a mild influence on gut microbes, thus resulting in the disappearance or reduction of CDI recurrence. CDBN-YGXZ displayed potent activity against C. difficile in vitro and in vivo, reducing or preventing relapse in infected animals, which could merit further development as a potential drug candidate for treating CDI.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Cricetinae , Animales , Ratones , Vancomicina/farmacología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/prevención & control , Recurrencia
9.
J Chemother ; 35(6): 527-538, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36548909

RESUMEN

Gastric cancer (GC) is one of the most malignant tumors with high incidence and poor prognosis. Currently, the combination of surgery with chemo- or radiotherapy is widely applied therapeutic strategy against GC. However, development of drug resistance severely limited the clinical application of chemotherapy. Small nucleolar RNA host gene 1 (SNHG1) has been reported to be frequently overexpressed in diverse human tumors. Yet, the biological roles and mechanisms of SNHG1 in chemoresistant GC remain unclear. Expressions of lncRNA and miRNA were detected by qRT-PCR. Responses of GC cells to Taxol treatments were evaluated by cell viability assay and apoptosis assay. Glucose metabolism rate was examined by glucose uptake and extracellular acidification rate (ECAR). The lncRNA-miRNA interaction was validated by RNA pull-down assay and luciferase assays. This study reports that expressions of SNHG1 were significantly elevated in patients with GC and gastric cancer cell lines. Silencing SNHG1 effectively suppressed GC cells migration and increased the Taxol sensitivity of GC cells. Moreover, we detected remarkedly upregulated SNHG1 expression and increased glucose metabolism in Taxol resistant cell line, MKN-45 TXR. Low glucose supply rendered Taxol resistant cells more susceptible to Taxol treatment compared with that from MKN-45 parental cells. Bioinformatical analysis, RNA pull-down and luciferase assays verified that SNHG1 functioned as a ceRNA of miR-216b-5p in GC cells. Consistently, we detected miR-216b-5p was significantly downregulated in GC tumor specimens and Taxol resistant GC cells. The hexokinase 2 (HK2), a glucose metabolism key enzyme, was predicted and validated as a direct target of miR-216b-5p in GC cells. Finally, restoration of miR-216b-5p in SNHG1-overexpressing MKN-45 TXR cells successfully overrode the SNHG1-promoted Taxol resistance through targeting the HK2-glycolysis axis. This study uncovered new biological roles and molecular mechanisms of the lncRNA-SNHG1-mediated Taxol resistance of gastric cancer, suggesting targeting the SNHG1-miR-216b-5p-HK2 axis could be a potentially therapeutic approach against chemoresistant gastric cancer.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Neoplasias Gástricas , Humanos , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glucosa , MicroARNs/genética , MicroARNs/metabolismo , Paclitaxel/farmacología , ARN Largo no Codificante/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética
10.
Nat Commun ; 13(1): 5282, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36075902

RESUMEN

Enteroviruses cause a number of medically relevant and widespread human diseases with no approved antiviral therapies currently available. Host-directed therapies present an enticing option for this diverse genus of viruses. We have previously identified the actin histidine methyltransferase SETD3 as a critical host factor physically interacting with the viral protease 2A. Here, we report the 3.5 Å cryo-EM structure of SETD3 interacting with coxsackievirus B3 2A at two distinct interfaces, including the substrate-binding surface within the SET domain. Structure-function analysis revealed that mutations of key residues in the SET domain resulted in severely reduced binding to 2A and complete protection from enteroviral infection. Our findings provide insight into the molecular basis of the SETD3-2A interaction and a framework for the rational design of host-directed therapeutics against enteroviruses.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Antígenos Virales/metabolismo , Endopeptidasas/metabolismo , Enterovirus/genética , Histona Metiltransferasas/metabolismo , Humanos , Péptido Hidrolasas/metabolismo
11.
Cancer Manag Res ; 14: 2235-2241, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35909650

RESUMEN

Large cell neuroendocrine carcinoma (LCNEC) is a rare histological subtype of ovarian cancer. A few cases have been reported in the literature with extreme invasiveness and a poor prognosis. However, there still have not been accepted criteria for diagnosis and treatment of LCNEC. Here we report an unmarried 37 year-old woman who was diagnosed with LCNEC associated with clear cell carcinoma and the tumor index was manifested with a specific increase of AFP. The case received six courses of etoposide and carboplatin chemotherapy as an adjuvant therapy after primary curative surgery. However, she relapsed within 6 months after surgery and metastasized rapidly to distant organs despite combined chemotherapy of paclitaxel, cisplatin, and bevacizumab, and died 18 months after primary surgery. This is the first reported case of LCNEC manifested with a specific increase of AFP and characteristically metastasized to the spine as recurrence. Reviewing our case as well as previously reported cases, LCNEC present with aggressive malignancy and vulnerable to distant metastasis through a hematogenous approach, we conjectured that adding Bevacizumab in primary chemotherapy may be beneficial to extend disease-free survival. But so far there is no recommendation of this regimen for treatment of LCNEC in current guidelines. Further research is needed to confirm this view so as to find the best treatment of LCNEC and improve the prognosis of these patients.

12.
Pathol Res Pract ; 233: 153856, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35366462

RESUMEN

Dysregulation of the long noncoding RNA GAS5 in human cancer has been identified in recent studies. In this study, we confirmed a negative correlation between the GAS5 expression level and papillary thyroid carcinoma clinicopathologic characteristics, such as the tumor size, lymph node metastasis, the TNM stage and BRAFV600E mutation. The viability and metastasis of papillary thyroid carcinoma cells were detected by CCK-8 and transwell assays, respectively. The results showed that upregulation of GAS5 significantly inhibited papillary thyroid cancer cell growth, migration and invasion in vitro. RNA transcriptome sequencing was performed to explore the underlying targets of GAS5. Through qRT-PCR and Western blot analysis, we found that ectopic expression of GAS5 significantly increased IFI44 and STAT1 levels. Taken together, these findings suggest that GAS5 is a tumor suppressor in papillary thyroid carcinoma, and the action of GAS5 may be mediated through the IFNγ/STAT1 signaling pathway.


Asunto(s)
ARN Largo no Codificante , Neoplasias de la Tiroides , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/genética , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética
13.
Front Oncol ; 12: 848406, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35392234

RESUMEN

In spite of improvements in diagnostics and treatment of gastric cancer (GC), it remains the most common malignancy of human digestive system. It is now widely appreciated that long noncoding RNAs (lncRNAs) exert extensive regulatory effects on a spectrum of fundamental biological processes through diverse mechanisms. In this study, we explored the expression level and functional role of lncRNA RP11-138J23.1 in GC. Through bioinformatics analyses and in situ hybridization (ISH), we identified that RP11-138J23.1 was upregulated in GC tissue. Further study showed that RP11-138J23.1 knockdown significantly inhibited cell proliferation and metastatic ability. Whereas, RP11-138J23.1 overexpression could promote tumor cell growth and metastasis in vitro. Additionally, loss-of-function assays were used to confirm the role of RP11-138J23.1 in vivo. Mechanistically, RP11-138J23.1 exerted its oncogenic functions by binding to HuR protein and increasing stability of VAV3 mRNA. Overall, our study highlights the essential role of RP11-138J23.1 in GC, suggesting that RP11-138J23.1 might be a potent therapeutic target for patients with GC.

14.
Nature ; 602(7895): 129-134, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35082446

RESUMEN

Differentiation proceeds along a continuum of increasingly fate-restricted intermediates, referred to as canalization1,2. Canalization is essential for stabilizing cell fate, but the mechanisms that underlie robust canalization are unclear. Here we show that the BRG1/BRM-associated factor (BAF) chromatin-remodelling complex ATPase gene Brm safeguards cell identity during directed cardiogenesis of mouse embryonic stem cells. Despite the establishment of a well-differentiated precardiac mesoderm, Brm-/- cells predominantly became neural precursors, violating germ layer assignment. Trajectory inference showed a sudden acquisition of a non-mesodermal identity in Brm-/- cells. Mechanistically, the loss of Brm prevented de novo accessibility of primed cardiac enhancers while increasing the expression of neurogenic factor POU3F1, preventing the binding of the neural suppressor REST and shifting the composition of BRG1 complexes. The identity switch caused by the Brm mutation was overcome by increasing BMP4 levels during mesoderm induction. Mathematical modelling supports these observations and demonstrates that Brm deletion affects cell fate trajectory by modifying saddle-node bifurcations2. In the mouse embryo, Brm deletion exacerbated mesoderm-deleted Brg1-mutant phenotypes, severely compromising cardiogenesis, and reveals an in vivo role for Brm. Our results show that Brm is a compensable safeguard of the fidelity of mesoderm chromatin states, and support a model in which developmental canalization is not a rigid irreversible path, but a highly plastic trajectory.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Mesodermo/citología , Mesodermo/metabolismo , Miocitos Cardíacos/citología , Factores de Transcripción/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , ADN Helicasas/metabolismo , Embrión de Mamíferos , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Miocardio/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Factor 6 de Transcripción de Unión a Octámeros/metabolismo , Fenotipo , Proteínas Represoras/metabolismo , Células Madre/citología , Factores de Tiempo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
15.
Nature ; 599(7883): 152-157, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34646016

RESUMEN

Molecular switch proteins whose cycling between states is controlled by opposing regulators1,2 are central to biological signal transduction. As switch proteins function within highly connected interaction networks3, the fundamental question arises of how functional specificity is achieved when different processes share common regulators. Here we show that functional specificity of the small GTPase switch protein Gsp1 in Saccharomyces cerevisiae (the homologue of the human protein RAN)4 is linked to differential sensitivity of biological processes to different kinetics of the Gsp1 (RAN) switch cycle. We make 55 targeted point mutations to individual protein interaction interfaces of Gsp1 (RAN) and show through quantitative genetic5 and physical interaction mapping that Gsp1 (RAN) interface perturbations have widespread cellular consequences. Contrary to expectation, the cellular effects of the interface mutations group by their biophysical effects on kinetic parameters of the GTPase switch cycle and not by the targeted interfaces. Instead, we show that interface mutations allosterically tune the GTPase cycle kinetics. These results suggest a model in which protein partner binding, or post-translational modifications at distal sites, could act as allosteric regulators of GTPase switching. Similar mechanisms may underlie regulation by other GTPases, and other biological switches. Furthermore, our integrative platform to determine the quantitative consequences of molecular perturbations may help to explain the effects of disease mutations that target central molecular switches.


Asunto(s)
Regulación Alostérica/genética , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mutación Puntual , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae , Sitios de Unión/genética , Dominio Catalítico/genética , Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanosina Trifosfato/metabolismo , Cinética , Unión Proteica/genética , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética
16.
PLoS Biol ; 19(4): e3001191, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33886552

RESUMEN

The Hedgehog (Hh) pathway is essential for organ development, homeostasis, and regeneration. Dysfunction of this cascade drives several cancers. To control expression of pathway target genes, the G protein-coupled receptor (GPCR) Smoothened (SMO) activates glioma-associated (GLI) transcription factors via an unknown mechanism. Here, we show that, rather than conforming to traditional GPCR signaling paradigms, SMO activates GLI by binding and sequestering protein kinase A (PKA) catalytic subunits at the membrane. This sequestration, triggered by GPCR kinase (GRK)-mediated phosphorylation of SMO intracellular domains, prevents PKA from phosphorylating soluble substrates, releasing GLI from PKA-mediated inhibition. Our work provides a mechanism directly linking Hh signal transduction at the membrane to GLI transcription in the nucleus. This process is more fundamentally similar between species than prevailing hypotheses suggest. The mechanism described here may apply broadly to other GPCR- and PKA-containing cascades in diverse areas of biology.


Asunto(s)
Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Hedgehog/metabolismo , Receptor Smoothened/fisiología , Animales , Animales Modificados Genéticamente , Dominio Catalítico/genética , Células Cultivadas , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/química , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Embrión no Mamífero , Células HEK293 , Proteínas Hedgehog/genética , Humanos , Ratones , Dominios y Motivos de Interacción de Proteínas/genética , Transducción de Señal/genética , Receptor Smoothened/metabolismo , Pez Cebra
17.
Science ; 370(6522)2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33303586

RESUMEN

Determining structures of protein complexes is crucial for understanding cellular functions. Here, we describe an integrative structure determination approach that relies on in vivo measurements of genetic interactions. We construct phenotypic profiles for point mutations crossed against gene deletions or exposed to environmental perturbations, followed by converting similarities between two profiles into an upper bound on the distance between the mutated residues. We determine the structure of the yeast histone H3-H4 complex based on ~500,000 genetic interactions of 350 mutants. We then apply the method to subunits Rpb1-Rpb2 of yeast RNA polymerase II and subunits RpoB-RpoC of bacterial RNA polymerase. The accuracy is comparable to that based on chemical cross-links; using restraints from both genetic interactions and cross-links further improves model accuracy and precision. The approach provides an efficient means to augment integrative structure determination with in vivo observations.


Asunto(s)
Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Mapas de Interacción de Proteínas/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Histonas/química , Histonas/genética , Mutación , Conformación Proteica , Mapeo de Interacción de Proteínas , Saccharomyces cerevisiae/genética
18.
J Int Med Res ; 48(11): 300060520971495, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33233994

RESUMEN

We present the case of a woman diagnosed with a teratoma adherent to the vaginal wall. The patient had been misdiagnosed with an ovarian teratoma 8 years previously at her local hospital, but no mass was found in the pelvic cavity during cesarean section. She therefore attended our institution for further examination. Transvaginal ultrasonography, magnetic resonance imaging (MRI), and computed tomography (CT) revealed a large mass on the left side at the bottom of the pelvis, near the side of the vagina, mainly composed of greasy and cystic elements. Gynecological examination showed the mass protruding into the left side of the vaginal wall. The patient therefore underwent vaginal wall incision. During surgery, we found a mass adherent to the vaginal wall, located on the left front of the rectum. Surgery was completed successful with no complications. This case highlights the need for careful preoperative evaluation of teratomas with unusual locations. MRI and CT may be useful for identifying the origin of the tumor and determining its relationship with the surrounding tissues. Surgery should be based on the characteristics and anatomical location of the tumor to minimize damage to other tissues and organs.


Asunto(s)
Quiste Dermoide , Neoplasias Ováricas , Teratoma , Cesárea , Femenino , Humanos , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/cirugía , Embarazo , Teratoma/diagnóstico por imagen , Teratoma/cirugía , Vagina/diagnóstico por imagen , Vagina/cirugía
19.
Science ; 370(6521)2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33060197

RESUMEN

The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a grave threat to public health and the global economy. SARS-CoV-2 is closely related to the more lethal but less transmissible coronaviruses SARS-CoV-1 and Middle East respiratory syndrome coronavirus (MERS-CoV). Here, we have carried out comparative viral-human protein-protein interaction and viral protein localization analyses for all three viruses. Subsequent functional genetic screening identified host factors that functionally impinge on coronavirus proliferation, including Tom70, a mitochondrial chaperone protein that interacts with both SARS-CoV-1 and SARS-CoV-2 ORF9b, an interaction we structurally characterized using cryo-electron microscopy. Combining genetically validated host factors with both COVID-19 patient genetic data and medical billing records identified molecular mechanisms and potential drug treatments that merit further molecular and clinical study.


Asunto(s)
COVID-19/metabolismo , Proteínas de la Nucleocápside de Coronavirus/metabolismo , Interacciones Microbiota-Huesped , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Mapas de Interacción de Proteínas , SARS-CoV-2/metabolismo , Síndrome Respiratorio Agudo Grave/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Secuencia Conservada , Proteínas de la Nucleocápside de Coronavirus/genética , Microscopía por Crioelectrón , Humanos , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Conformación Proteica
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA