Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.138
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38701782

RESUMEN

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Asunto(s)
Epigénesis Genética , Vaina de Mielina , Oligodendroglía , Remielinización , Animales , Vaina de Mielina/metabolismo , Humanos , Ratones , Remielinización/efectos de los fármacos , Oligodendroglía/metabolismo , Sistema Nervioso Central/metabolismo , Ratones Endogámicos C57BL , Rejuvenecimiento , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Organoides/metabolismo , Organoides/efectos de los fármacos , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/genética , Diferenciación Celular/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Masculino , Regeneración/efectos de los fármacos , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología
2.
Nat Immunol ; 24(4): 625-636, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36941398

RESUMEN

The intestinal immune system interacts with commensal microbiota to maintain gut homeostasis. Furthermore, stress alters the microbiome composition, leading to impaired brain function; yet how the intestinal immune system mediates these effects remains elusive. Here we report that colonic γδ T cells modulate behavioral vulnerability to chronic social stress via dectin-1 signaling. We show that reduction in specific Lactobacillus species, which are involved in T cell differentiation to protect the host immune system, contributes to stress-induced social-avoidance behavior, consistent with our observations in patients with depression. Stress-susceptible behaviors derive from increased differentiation in colonic interleukin (IL)-17-producing γδ T cells (γδ17 T cells) and their meningeal accumulation. These stress-susceptible cellular and behavioral phenotypes are causally mediated by dectin-1, an innate immune receptor expressed in γδ T cells. Our results highlight the previously unrecognized role of intestinal γδ17 T cells in the modulation of psychological stress responses and the importance of dectin-1 as a potential therapeutic target for the treatment of stress-induced behaviors.


Asunto(s)
Intestinos , Lectinas Tipo C , Colon , Transducción de Señal , Receptores de Antígenos de Linfocitos T gamma-delta
3.
Nature ; 614(7946): 102-107, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36697827

RESUMEN

Living amphibians (Lissamphibia) include frogs and salamanders (Batrachia) and the limbless worm-like caecilians (Gymnophiona). The estimated Palaeozoic era gymnophionan-batrachian molecular divergence1 suggests a major gap in the record of crown lissamphibians prior to their earliest fossil occurrences in the Triassic period2-6. Recent studies find a monophyletic Batrachia within dissorophoid temnospondyls7-10, but the absence of pre-Jurassic period caecilian fossils11,12 has made their relationships to batrachians and affinities to Palaeozoic tetrapods controversial1,8,13,14. Here we report the geologically oldest stem caecilian-a crown lissamphibian from the Late Triassic epoch of Arizona, USA-extending the caecilian record by around 35 million years. These fossils illuminate the tempo and mode of early caecilian morphological and functional evolution, demonstrating a delayed acquisition of musculoskeletal features associated with fossoriality in living caecilians, including the dual jaw closure mechanism15,16, reduced orbits17 and the tentacular organ18. The provenance of these fossils suggests a Pangaean equatorial origin for caecilians, implying that living caecilian biogeography reflects conserved aspects of caecilian function and physiology19, in combination with vicariance patterns driven by plate tectonics20. These fossils reveal a combination of features that is unique to caecilians alongside features that are shared with batrachian and dissorophoid temnospondyls, providing new and compelling evidence supporting a single origin of living amphibians within dissorophoid temnospondyls.


Asunto(s)
Anfibios , Anuros , Fósiles , Filogenia , Urodelos , Animales , Anfibios/anatomía & histología , Anuros/anatomía & histología , Arizona , Urodelos/anatomía & histología , Órbita/anatomía & histología , Maxilares/anatomía & histología , Sistema Musculoesquelético/anatomía & histología
4.
Nature ; 619(7968): 135-142, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37316671

RESUMEN

Vaccination with Sabin, a live attenuated oral polio vaccine (OPV), results in robust intestinal and humoral immunity and has been key to controlling poliomyelitis. As with any RNA virus, OPV evolves rapidly to lose attenuating determinants critical to the reacquisition of virulence1-3 resulting in vaccine-derived, virulent poliovirus variants. Circulation of these variants within underimmunized populations leads to further evolution of circulating, vaccine-derived poliovirus with higher transmission capacity, representing a significant risk of polio re-emergence. A new type 2 OPV (nOPV2), with promising clinical data on genetic stability and immunogenicity, recently received authorization from the World Health Organization for use in response to circulating, vaccine-derived poliovirus outbreaks. Here we report the development of two additional live attenuated vaccine candidates against type 1 and 3 polioviruses. The candidates were generated by replacing the capsid coding region of nOPV2 with that from Sabin 1 or 3. These chimeric viruses show growth phenotypes similar to nOPV2 and immunogenicity comparable to their parental Sabin strains, but are more attenuated. Our experiments in mice and deep sequencing analysis confirmed that the candidates remain attenuated and preserve all the documented nOPV2 characteristics concerning genetic stability following accelerated virus evolution. Importantly, these vaccine candidates are highly immunogenic in mice as monovalent and multivalent formulations and may contribute to poliovirus eradication.


Asunto(s)
Poliomielitis , Vacuna Antipolio Oral , Poliovirus , Vacunas Atenuadas , Animales , Ratones , Modelos Animales de Enfermedad , Poliomielitis/inmunología , Poliomielitis/prevención & control , Poliomielitis/virología , Poliovirus/clasificación , Poliovirus/genética , Poliovirus/inmunología , Vacuna Antipolio Oral/química , Vacuna Antipolio Oral/genética , Vacuna Antipolio Oral/inmunología , Vacunas Atenuadas/química , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Erradicación de la Enfermedad
5.
Cell ; 152(5): 1008-20, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23452850

RESUMEN

Metazoan evolution involves increasing protein domain complexity, but how this relates to control of biological decisions remains uncertain. The Ras guanine nucleotide exchange factor (RasGEF) Sos1 and its adaptor Grb2 are multidomain proteins that couple fibroblast growth factor (FGF) signaling to activation of the Ras-Erk pathway during mammalian development and drive embryonic stem cells toward the primitive endoderm (PrE) lineage. We show that the ability of Sos1/Grb2 to appropriately regulate pluripotency and differentiation factors and to initiate PrE development requires collective binding of multiple Sos1/Grb2 domains to their protein and phospholipid ligands. This provides a cooperative system that only allows lineage commitment when all ligand-binding domains are occupied. Furthermore, our results indicate that the interaction domains of Sos1 and Grb2 have evolved so as to bind ligands not with maximal strength but with specificities and affinities that maintain cooperativity. This optimized system ensures that PrE lineage commitment occurs in a timely and selective manner during embryogenesis.


Asunto(s)
Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteína SOS1/metabolismo , Secuencia de Aminoácidos , Animales , Linaje de la Célula , Endodermo/metabolismo , Eucariontes/genética , Eucariontes/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Factores de Intercambio de Guanina Nucleótido ras/metabolismo
6.
Nature ; 597(7878): 709-714, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497421

RESUMEN

Multiple sclerosis (MS) lesions that do not resolve in the months after they form harbour ongoing demyelination and axon degeneration, and are identifiable in vivo by their paramagnetic rims on MRI scans1-3. Here, to define mechanisms underlying this disabling, progressive neurodegenerative state4-6 and foster development of new therapeutic agents, we used MRI-informed single-nucleus RNA sequencing to profile the edge of demyelinated white matter lesions at various stages of inflammation. We uncovered notable glial and immune cell diversity, especially at the chronically inflamed lesion edge. We define 'microglia inflamed in MS' (MIMS) and 'astrocytes inflamed in MS', glial phenotypes that demonstrate neurodegenerative programming. The MIMS transcriptional profile overlaps with that of microglia in other neurodegenerative diseases, suggesting that primary and secondary neurodegeneration share common mechanisms and could benefit from similar therapeutic approaches. We identify complement component 1q (C1q) as a critical mediator of MIMS activation, validated immunohistochemically in MS tissue, genetically by microglia-specific C1q ablation in mice with experimental autoimmune encephalomyelitis, and therapeutically by treating chronic experimental autoimmune encephalomyelitis with C1q blockade. C1q inhibition is a potential therapeutic avenue to address chronic white matter inflammation, which could be monitored by longitudinal assessment of its dynamic biomarker, paramagnetic rim lesions, using advanced MRI methods.


Asunto(s)
Astrocitos/patología , Linfocitos/patología , Microglía/patología , Esclerosis Múltiple/patología , Animales , Encéfalo/patología , Complemento C1q/antagonistas & inhibidores , Complemento C1q/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inflamación/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Esclerosis Múltiple/diagnóstico por imagen , RNA-Seq , Transcriptoma , Sustancia Blanca/patología
7.
Proc Natl Acad Sci U S A ; 121(11): e2311726121, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38451939

RESUMEN

Proteins are a diverse class of biomolecules responsible for wide-ranging cellular functions, from catalyzing reactions to recognizing pathogens. The ability to evolve proteins rapidly and inexpensively toward improved properties is a common objective for protein engineers. Powerful high-throughput methods like fluorescent activated cell sorting and next-generation sequencing have dramatically improved directed evolution experiments. However, it is unclear how to best leverage these data to characterize protein fitness landscapes more completely and identify lead candidates. In this work, we develop a simple yet powerful framework to improve protein optimization by predicting continuous protein properties from simple directed evolution experiments using interpretable, linear machine learning models. Importantly, we find that these models, which use data from simple but imprecise experimental estimates of protein fitness, have predictive capabilities that approach more precise but expensive data. Evaluated across five diverse protein engineering tasks, continuous properties are consistently predicted from readily available deep sequencing data, demonstrating that protein fitness space can be reasonably well modeled by linear relationships among sequence mutations. To prospectively test the utility of this approach, we generated a library of stapled peptides and applied the framework to predict affinity and specificity from simple cell sorting data. We then coupled integer linear programming, a method to optimize protein fitness from linear weights, with mutation scores from machine learning to identify variants in unseen sequence space that have improved and co-optimal properties. This approach represents a versatile tool for improved analysis and identification of protein variants across many domains of protein engineering.


Asunto(s)
Aprendizaje Automático , Proteínas , Proteínas/metabolismo , Ingeniería de Proteínas/métodos , Mutación , Biblioteca de Genes
8.
J Cell Sci ; 137(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38606629

RESUMEN

The ADP-ribosylation factors (ARFs) and ARF-like (ARL) GTPases serve as essential molecular switches governing a wide array of cellular processes. In this study, we used proximity-dependent biotin identification (BioID) to comprehensively map the interactome of 28 out of 29 ARF and ARL proteins in two cellular models. Through this approach, we identified ∼3000 high-confidence proximal interactors, enabling us to assign subcellular localizations to the family members. Notably, we uncovered previously undefined localizations for ARL4D and ARL10. Clustering analyses further exposed the distinctiveness of the interactors identified with these two GTPases. We also reveal that the expression of the understudied member ARL14 is confined to the stomach and intestines. We identified phospholipase D1 (PLD1) and the ESCPE-1 complex, more precisely, SNX1, as proximity interactors. Functional assays demonstrated that ARL14 can activate PLD1 in cellulo and is involved in cargo trafficking via the ESCPE-1 complex. Overall, the BioID data generated in this study provide a valuable resource for dissecting the complexities of ARF and ARL spatial organization and signaling.


Asunto(s)
Factores de Ribosilacion-ADP , Fosfolipasa D , Transducción de Señal , Factores de Ribosilacion-ADP/metabolismo , Factores de Ribosilacion-ADP/genética , Humanos , Fosfolipasa D/metabolismo , Fosfolipasa D/genética , Células HEK293 , Animales , Nexinas de Clasificación/metabolismo , Nexinas de Clasificación/genética , Mapeo de Interacción de Proteínas
9.
PLoS Biol ; 21(4): e3002068, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37011096

RESUMEN

Given the requisite cost associated with observing species interactions, ecologists often reuse species interaction networks created by different sets of researchers to test their hypotheses regarding how ecological processes drive network topology. Yet, topological properties identified across these networks may not be sufficiently attributable to ecological processes alone as often assumed. Instead, much of the totality of topological differences between networks-topological heterogeneity-could be due to variations in research designs and approaches that different researchers use to create each species interaction network. To evaluate the degree to which this topological heterogeneity is present in available ecological networks, we first compared the amount of topological heterogeneity across 723 species interaction networks created by different sets of researchers with the amount quantified from non-ecological networks known to be constructed following more consistent approaches. Then, to further test whether the topological heterogeneity was due to differences in study designs, and not only to inherent variation within ecological networks, we compared the amount of topological heterogeneity between species interaction networks created by the same sets of researchers (i.e., networks from the same publication) with the amount quantified between networks that were each from a unique publication source. We found that species interaction networks are highly topologically heterogeneous: while species interaction networks from the same publication are much more topologically similar to each other than interaction networks that are from a unique publication, they still show at least twice as much heterogeneity as any category of non-ecological networks that we tested. Altogether, our findings suggest that extra care is necessary to effectively analyze species interaction networks created by different researchers, perhaps by controlling for the publication source of each network.

10.
Circ Res ; 134(10): 1306-1326, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38533639

RESUMEN

BACKGROUND: Ventricular arrhythmias (VAs) demonstrate a prominent day-night rhythm, commonly presenting in the morning. Transcriptional rhythms in cardiac ion channels accompany this phenomenon, but their role in the morning vulnerability to VAs and the underlying mechanisms are not understood. We investigated the recruitment of transcription factors that underpins transcriptional rhythms in ion channels and assessed whether this mechanism was pertinent to the heart's intrinsic diurnal susceptibility to VA. METHODS AND RESULTS: Assay for transposase-accessible chromatin with sequencing performed in mouse ventricular myocyte nuclei at the beginning of the animals' inactive (ZT0) and active (ZT12) periods revealed differentially accessible chromatin sites annotating to rhythmically transcribed ion channels and distinct transcription factor binding motifs in these regions. Notably, motif enrichment for the glucocorticoid receptor (GR; transcriptional effector of corticosteroid signaling) in open chromatin profiles at ZT12 was observed, in line with the well-recognized ZT12 peak in circulating corticosteroids. Molecular, electrophysiological, and in silico biophysically-detailed modeling approaches demonstrated GR-mediated transcriptional control of ion channels (including Scn5a underlying the cardiac Na+ current, Kcnh2 underlying the rapid delayed rectifier K+ current, and Gja1 responsible for electrical coupling) and their contribution to the day-night rhythm in the vulnerability to VA. Strikingly, both pharmacological block of GR and cardiomyocyte-specific genetic knockout of GR blunted or abolished ion channel expression rhythms and abolished the ZT12 susceptibility to pacing-induced VA in isolated hearts. CONCLUSIONS: Our study registers a day-night rhythm in chromatin accessibility that accompanies diurnal cycles in ventricular myocytes. Our approaches directly implicate the cardiac GR in the myocyte excitability rhythm and mechanistically link the ZT12 surge in glucocorticoids to intrinsic VA propensity at this time.


Asunto(s)
Ritmo Circadiano , Miocitos Cardíacos , Receptores de Glucocorticoides , Animales , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Ratones , Miocitos Cardíacos/metabolismo , Masculino , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/genética , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Conexina 43/metabolismo , Conexina 43/genética , Ratones Noqueados , Potenciales de Acción
11.
EMBO Rep ; 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009833

RESUMEN

RAS GTPases bind effectors to convert upstream cues to changes in cellular function. Effectors of classical H/K/NRAS are defined by RBD/RA domains which recognize the GTP-bound conformation of these GTPases, yet the specificity of RBD/RAs for over 160 RAS superfamily proteins remains poorly explored. We have systematically mapped interactions between BRAF and four RASSF effectors, the largest family of RA-containing proteins, with all RAS, RHO and ARF small GTPases. 39 validated complexes reveal plasticity in RASSF binding, while BRAF demonstrates tight specificity for classical H/K/NRAS. Complex between RASSF5 and diverse RAS GTPases at the plasma membrane can activate Hippo signalling and sequester YAP in the cytosol. RASSF8 undergoes liquid-liquid phase separation and resides in YAP-associated membraneless condensates, which also engage several RAS and RHO GTPases. The poorly studied RASSF3 has been identified as a first potential effector of mitochondrial MIRO proteins, and its co-expression with these GTPases impacts mitochondria and peroxisome distribution. These data reveal the complex nature of GTPase-effector interactions and show their systematic elucidation can reveal completely novel and biologically relevant cellular processes.

12.
J Immunol ; 212(4): 563-575, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38149923

RESUMEN

Patients infected with SARS-CoV-2 experience variable disease susceptibility, and patients with comorbidities such as sepsis are often hospitalized for COVID-19 complications. However, the extent to which initial infectious inoculum dose determines disease outcomes and whether this can be used for immunological priming in a genetically susceptible host has not been completely defined. We used an established SARS-like murine model in which responses to primary and/or secondary challenges with murine hepatitis virus type 1 (MHV-1) were analyzed. We compared the response to infection in genetically susceptible C3H/HeJ mice, genetically resistant C57BL/6J mice, and genetically diverse, variably susceptible outbred Swiss Webster mice. Although defined as genetically susceptible to MHV-1, C3H/HeJ mice displayed decreasing dose-dependent pathological changes in disease severity and lung infiltrate/edema, as well as lymphopenia. Importantly, an asymptomatic dose (500 PFU) was identified that yielded no measurable morbidity/mortality postinfection in C3H/HeJ mice. Polymicrobial sepsis induced via cecal ligation and puncture converted asymptomatic infections in C3H/HeJ and C57BL/6J mice to more pronounced disease, modeling the impact of sepsis as a comorbidity to ß-coronavirus infection. We then used low-dose infection as an immunological priming event in C3H/HeJ mice, which provided neutralizing Ab-dependent, but not circulating CD4/CD8 T cell-dependent, protection against a high-dose MHV-1 early rechallenge. Together, these data define how infection dose, immunological status, and comorbidities modulate outcomes of primary and secondary ß-coronavirus infections in hosts with variable susceptibility.


Asunto(s)
Virus de la Hepatitis Murina , Sepsis , Humanos , Ratones , Animales , Ratones Endogámicos C57BL , Ratones Endogámicos C3H , Ratones Endogámicos , Predisposición Genética a la Enfermedad
13.
Proc Natl Acad Sci U S A ; 120(28): e2219543120, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37406092

RESUMEN

Although HIV-1 Gag is known to drive viral assembly and budding, the precise mechanisms by which the lipid composition of the plasma membrane is remodeled during assembly are incompletely understood. Here, we provide evidence that the sphingomyelin hydrolase neutral sphingomyelinase 2 (nSMase2) interacts with HIV-1 Gag and through the hydrolysis of sphingomyelin creates ceramide that is necessary for proper formation of the viral envelope and viral maturation. Inhibition or depletion of nSMase2 resulted in the production of noninfectious HIV-1 virions with incomplete Gag lattices lacking condensed conical cores. Inhibition of nSMase2 in HIV-1-infected humanized mouse models with a potent and selective inhibitor of nSMase2 termed PDDC [phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2, 6-dimethylimidazo[1,2-b]pyridazin-8-yl) pyrrolidin-3-yl)-carbamate] produced a linear reduction in levels of HIV-1 in plasma. If undetectable plasma levels of HIV-1 were achieved with PDDC treatment, viral rebound did not occur for up to 4 wk when PDDC was discontinued. In vivo and tissue culture results suggest that PDDC selectively kills cells with actively replicating HIV-1. Collectively, this work demonstrates that nSMase2 is a critical regulator of HIV-1 replication and suggests that nSMase2 could be an important therapeutic target with the potential to kill HIV-1-infected cells.


Asunto(s)
VIH-1 , Esfingomielina Fosfodiesterasa , Ratones , Animales , Esfingomielina Fosfodiesterasa/metabolismo , VIH-1/metabolismo , Esfingomielinas/metabolismo , Membrana Celular/metabolismo
14.
N Engl J Med ; 386(12): 1132-1142, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-35179323

RESUMEN

BACKGROUND: Darolutamide is a potent androgen-receptor inhibitor that has been associated with increased overall survival among patients with nonmetastatic, castration-resistant prostate cancer. Whether a combination of darolutamide, androgen-deprivation therapy, and docetaxel would increase survival among patients with metastatic, hormone-sensitive prostate cancer is unknown. METHODS: In this international, phase 3 trial, we randomly assigned patients with metastatic, hormone-sensitive prostate cancer in a 1:1 ratio to receive darolutamide (at a dose of 600 mg [two 300-mg tablets] twice daily) or matching placebo, both in combination with androgen-deprivation therapy and docetaxel. The primary end point was overall survival. RESULTS: The primary analysis involved 1306 patients (651 in the darolutamide group and 655 in the placebo group); 86.1% of the patients had disease that was metastatic at the time of the initial diagnosis. At the data cutoff date for the primary analysis (October 25, 2021), the risk of death was significantly lower, by 32.5%, in the darolutamide group than in the placebo group (hazard ratio 0.68; 95% confidence interval, 0.57 to 0.80; P<0.001). Darolutamide was also associated with consistent benefits with respect to the secondary end points and prespecified subgroups. Adverse events were similar in the two groups, and the incidences of the most common adverse events (occurring in ≥10% of the patients) were highest during the overlapping docetaxel treatment period in both groups. The frequency of grade 3 or 4 adverse events was 66.1% in the darolutamide group and 63.5% in the placebo group; neutropenia was the most common grade 3 or 4 adverse event (in 33.7% and 34.2%, respectively). CONCLUSIONS: In this trial involving patients with metastatic, hormone-sensitive prostate cancer, overall survival was significantly longer with the combination of darolutamide, androgen-deprivation therapy, and docetaxel than with placebo plus androgen-deprivation therapy and docetaxel, and the addition of darolutamide led to improvement in key secondary end points. The frequency of adverse events was similar in the two groups. (Funded by Bayer and Orion Pharma; ARASENS ClinicalTrials.gov number, NCT02799602.).


Asunto(s)
Antagonistas de Receptores Androgénicos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Pirazoles/uso terapéutico , Anciano , Anciano de 80 o más Años , Antagonistas de Andrógenos/uso terapéutico , Antagonistas de Receptores Androgénicos/efectos adversos , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Docetaxel/efectos adversos , Docetaxel/uso terapéutico , Quimioterapia Combinada , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia/tratamiento farmacológico , Neutropenia/inducido químicamente , Modelos de Riesgos Proporcionales , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración , Pirazoles/efectos adversos
15.
PLoS Comput Biol ; 20(7): e1012258, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38968291

RESUMEN

The practical application of new single molecule protein sequencing (SMPS) technologies requires accurate estimates of their associated sequencing error rates. Here, we describe the development and application of two distinct parameter estimation methods for analyzing SMPS reads produced by fluorosequencing. A Hidden Markov Model (HMM) based approach, extends whatprot, where we previously used HMMs for SMPS peptide-read matching. This extension offers a principled approach for estimating key parameters for fluorosequencing experiments, including missed amino acid cleavages, dye loss, and peptide detachment. Specifically, we adapted the Baum-Welch algorithm, a standard technique to estimate transition probabilities for an HMM using expectation maximization, but modified here to estimate a small number of parameter values directly rather than estimating every transition probability independently. We demonstrate a high degree of accuracy on simulated data, but on experimental datasets, we observed that the model needed to be augmented with an additional error type, N-terminal blocking. This, in combination with data pre-processing, results in reasonable parameterizations of experimental datasets that agree with controlled experimental perturbations. A second independent implementation using a hybrid of DIRECT and Powell's method to reduce the root mean squared error (RMSE) between simulations and the real dataset was also developed. We compare these methods on both simulated and real data, finding that our Baum-Welch based approach outperforms DIRECT and Powell's method by most, but not all, criteria. Although some discrepancies between the results exist, we also find that both approaches provide similar error rate estimates from experimental single molecule fluorosequencing datasets.


Asunto(s)
Algoritmos , Cadenas de Markov , Análisis de Secuencia de Proteína , Análisis de Secuencia de Proteína/métodos , Proteínas/química , Biología Computacional/métodos , Imagen Individual de Molécula/métodos , Simulación por Computador
16.
Cell ; 141(1): 117-28, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20371349

RESUMEN

The association of p120 catenin (p120) with the juxtamembrane domain (JMD) of the cadherin cytoplasmic tail is critical for the surface stability of cadherin-catenin cell-cell adhesion complexes. Here, we present the crystal structure of p120 isoform 4A in complex with the JMD core region (JMD(core)) of E-cadherin. The p120 armadillo repeat domain contains modular binding pockets that are complementary to electrostatic and hydrophobic properties of the JMD(core). Single-residue mutations within the JMD(core)-binding site of p120 abolished its interaction with E- and N-cadherins in vitro and in cultured cells. These mutations of p120 enabled us to clearly differentiate between N-cadherin-dependent and -independent steps of neuronal dendritic spine morphogenesis crucial for synapse development. NMR studies revealed that p120 regulates the stability of cadherin-mediated cell-cell adhesion by associating with the majority of the JMD, including residues implicated in clathrin-mediated endocytosis and Hakai-dependent ubiquitination of E-cadherin, through its discrete "dynamic" and "static" binding sites.


Asunto(s)
Cadherinas/química , Cadherinas/metabolismo , Cateninas/química , Cateninas/metabolismo , Adhesión Celular , Animales , Cadherinas/genética , Cateninas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Ratones , Modelos Moleculares , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Catenina delta
17.
Bioessays ; 45(9): e2300088, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37401638

RESUMEN

RAS GTPases play essential roles in normal development and are direct drivers of human cancers. Three decades of study have failed to wholly characterize pathways stimulated by activated RAS, driven by engagement with 'effector' proteins that have RAS binding domains (RBDs). Bone fide effectors must bind directly to RAS GTPases in a nucleotide-dependent manner, and this interaction must impart a clear change in effector activity. Despite this, for most proteins currently deemed effectors there is little mechanistic understanding of how binding to the GTPase alters protein function. There has also been limited effort to comprehensively resolve the specificity of effector binding to the full array of RAS superfamily GTPase proteins. This review will summarize what is known about RAS-driven activation for an array of potential effector proteins, focusing on structural and mechanistic effects and highlighting how little is still known regarding this key paradigm of cellular signal transduction.


Asunto(s)
Transducción de Señal , Proteínas ras , Humanos , Proteínas ras/metabolismo , Unión Proteica , Proteínas/metabolismo , Nucleótidos/metabolismo
18.
Proc Natl Acad Sci U S A ; 119(26): e2121868119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35727980

RESUMEN

Proper orientation of the mitotic spindle plays a crucial role in embryos, during tissue development, and in adults, where it functions to dissipate mechanical stress to maintain tissue integrity and homeostasis. While mitotic spindles have been shown to reorient in response to external mechanical stresses, the subcellular cues that mediate spindle reorientation remain unclear. Here, we used a combination of optogenetics and computational modeling to investigate how mitotic spindles respond to inhomogeneous tension within the actomyosin cortex. Strikingly, we found that the optogenetic activation of RhoA only influences spindle orientation when it is induced at both poles of the cell. Under these conditions, the sudden local increase in cortical tension induced by RhoA activation reduces pulling forces exerted by cortical regulators on astral microtubules. This leads to a perturbation of the balance of torques exerted on the spindle, which causes it to rotate. Thus, spindle rotation in response to mechanical stress is an emergent phenomenon arising from the interaction between the spindle positioning machinery and the cell cortex.


Asunto(s)
Microtúbulos , Huso Acromático , Estrés Mecánico , Actomiosina/metabolismo , Simulación por Computador , Citoplasma , Microtúbulos/metabolismo , Optogenética , Huso Acromático/fisiología , Proteína de Unión al GTP rhoA/metabolismo
19.
J Allergy Clin Immunol ; 153(4): 1113-1124.e7, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38065233

RESUMEN

BACKGROUND: Patients with deleterious variants in MYSM1 have an immune deficiency characterized by B-cell lymphopenia, hypogammaglobulinemia, and increased radiosensitivity. MYSM1 is a histone deubiquitinase with established activity in regulating gene expression. MYSM1 also localizes to sites of DNA injury but its function in cellular responses to DNA breaks has not been elucidated. OBJECTIVES: This study sought to determine the activity of MYSM1 in regulating DNA damage responses (DDRs) to DNA double-stranded breaks (DSBs) generated during immunoglobulin receptor gene (Ig) recombination and by ionizing radiation. METHODS: MYSM1-deficient pre- and non-B cells were used to determine the role of MYSM1 in DSB generation, DSB repair, and termination of DDRs. RESULTS: Genetic testing in a newborn with abnormal screen for severe combined immune deficiency, T-cell lymphopenia, and near absence of B cells identified a novel splice variant in MYSM1 that results in nearly absent protein expression. Radiosensitivity testing in patient's peripheral blood lymphocytes showed constitutive γH2AX, a marker of DNA damage, in B cells in the absence of irradiation, suggesting a role for MYSM1 in response to DSBs generated during Ig recombination. Suppression of MYSM1 in pre-B cells did not alter generation or repair of Ig DSBs. Rather, loss of MYSM1 resulted in persistent DNA damage foci and prolonged DDR signaling. Loss of MYSM1 also led to protracted DDRs in U2OS cells with irradiation induced DSBs. CONCLUSIONS: MYSM1 regulates termination of DNA damage responses but does not function in DNA break generation and repair.


Asunto(s)
Daño del ADN , Reparación del ADN , Linfopenia , Humanos , Recién Nacido , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Histonas/genética , Histonas/metabolismo , Linfopenia/genética , Transactivadores/genética , Proteasas Ubiquitina-Específicas/genética , Proteasas Ubiquitina-Específicas/metabolismo
20.
J Biol Chem ; 299(9): 105123, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37536630

RESUMEN

Distinct functions mediated by members of the monopolar spindle-one-binder (MOB) family of proteins remain elusive beyond the evolutionarily conserved and well-established roles of MOB1 (MOB1A/B) in regulating tissue homeostasis within the Hippo pathway. Since MOB proteins are adaptors, understanding how they engage in protein-protein interactions and help assemble complexes is essential to define the full scope of their biological functions. To address this, we undertook a proximity-dependent biotin identification approach to define the interactomes of all seven human MOB proteins in HeLa and human embryonic kidney 293 cell lines. We uncovered >200 interactions, of which at least 70% are unreported on BioGrid. The generated dataset reliably recalled the bona fide interactors of the well-studied MOBs. We further defined the common and differential interactome between different MOBs on a subfamily and an individual level. We discovered a unique association between MOB3C and 7 of 10 protein subunits of the RNase P complex, an endonuclease that catalyzes tRNA 5' maturation. As a proof of principle for the robustness of the generated dataset, we validated the specific interaction of MOB3C with catalytically active RNase P by using affinity purification-mass spectrometry and pre-tRNA cleavage assays of MOB3C pulldowns. In summary, our data provide novel insights into the biology of MOB proteins and reveal the first interactors of MOB3C, components of the RNase P complex, and hence an exciting nexus with RNA biology.


Asunto(s)
Vía de Señalización Hippo , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas , Ribonucleasa P , Humanos , Células HeLa , Vía de Señalización Hippo/fisiología , Ribonucleasa P/metabolismo , Células HEK293 , Subunidades de Proteína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA