Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 616
Filtrar
1.
Biochemistry ; 60(45): 3385-3397, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34723504

RESUMEN

Condensins play a key role in higher order chromosome organization. In budding yeast Saccharomyces cerevisiae, a condensin complex consists of five subunits: two conserved structural maintenance of chromosome subunits, Smc2 and Smc4, a kleisin Brn1, and two HEAT repeat subunits, Ycg1, which possesses a DNA binding activity, and Ycs4, which can transiently associate with Smc4 and thereby disrupt its association with the Smc2 head. We characterized here DNA binding activity of the non-SMC subunits using an agnostic, model-independent approach. To this end, we mapped the DNA interface of the complex using sulfo-NHS biotin labeling. Besides the known site on Ycg1, we found a patch of lysines at the C-terminal domain of Ycs4 that were protected from biotinylation in the presence of DNA. Point mutations at the predicted protein-DNA interface reduced both Ycs4 binding to DNA and the DNA stimulated ATPase activity of the reconstituted condensin, whereas overproduction of the mutant Ycs4 was detrimental for yeast viability. Notably, the DNA binding site on Ycs4 partially overlapped with its interface with SMC4, revealing an intricate interplay between DNA binding, engagement of the Smc2-Smc4 heads, and ATP hydrolysis and suggesting a mechanism for ATP-modulated loading and translocation of condensins on DNA.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/fisiología , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , Adenosina Trifosfatasas/genética , Sitios de Unión/genética , Biotinilación , Comunicación Celular , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Complejos Multiproteicos/genética , Proteínas Nucleares , Fagocitosis , Mutación Puntual/genética , Dominios Proteicos/fisiología , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
2.
J Endocrinol ; 252(1): R23-R39, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34663757

RESUMEN

Ghrelin is a peptide hormone secreted primarily by the stomach that acts upon the growth hormone secretagogue receptor (GHSR1), a G protein-coupled receptor whose functions include growth hormone secretion, appetite regulation, energy expenditure, regulation of adiposity, and insulin release. Following the discovery that GHSR1a stimulates food intake, receptor antagonists were developed as potential therapies to regulate appetite. However, despite reductions in signalling, the desired effects on appetite were absent. Studies in the past 15 years have demonstrated GHSR1a can interact with other transmembrane proteins, either by direct binding (i.e. heteromerisation) or via signalling cross-talk. These interactions have various effects on GHSR1a signalling including preferential coupling to one pathway (i.e. biased signalling), coupling to a unique G protein (G protein switching), suppression of GHSR1a signalling, and enhancement of signalling by both receptors. While many of these interactions have been shown in cells overexpressing the proteins of interest and remain to be verified in tissues, substantial evidence exists showing that GHSR1a and the dopamine receptor D1 (DRD1) form heteromers, which promote synaptic plasticity and formation of hippocampal memory. Additionally, a reduction in GHSR1a-DRD1 complexes in favour of establishment of GHSR1a-Aß complexes correlates with Alzheimer's disease, indicating that GHSR1a heteromers may have pathological functions. Herein, we summarise the evidence published to date describing interactions between GHSR1a and transmembrane proteins, discuss the experimental strengths and limitations of these studies, describe the physiological evidence for each interaction, and address their potential as novel drug targets for appetite regulation, Alzheimer's disease, insulin secretion, and inflammation.


Asunto(s)
Complejos Multiproteicos/fisiología , Multimerización de Proteína/fisiología , Receptores de Ghrelina/fisiología , Animales , Ghrelina/metabolismo , Ghrelina/fisiología , Humanos , Complejos Multiproteicos/metabolismo , Unión Proteica/fisiología , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D1/fisiología , Receptores de Ghrelina/metabolismo , Transducción de Señal/fisiología
3.
Int J Mol Sci ; 22(15)2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34360575

RESUMEN

Many proteins have been found to operate in a complex with various biomolecules such as proteins, nucleic acids, carbohydrates, or lipids. Protein complexes can be transient, stable or dynamic and their association is controlled under variable cellular conditions. Complexome profiling is a recently developed mass spectrometry-based method that combines mild separation techniques, native gel electrophoresis, and density gradient centrifugation with quantitative mass spectrometry to generate inventories of protein assemblies within a cell or subcellular fraction. This review summarizes applications of complexome profiling with respect to assembly ranging from single subunits to large macromolecular complexes, as well as their stability, and remodeling in health and disease.


Asunto(s)
Complejos Multiproteicos/química , Complejos Multiproteicos/fisiología , Proteínas/química , Proteínas/fisiología , Animales , Humanos
4.
Mol Hum Reprod ; 27(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34191027

RESUMEN

Since its recent discovery, the subcortical maternal complex (SCMC) is emerging as a maternally inherited and crucial biological structure for the initial stages of embryogenesis in mammals. Uniquely expressed in oocytes and preimplantation embryos, where it localizes to the cell subcortex, this multiprotein complex is essential for early embryo development in the mouse and is functionally conserved across mammalian species, including humans. The complex has been linked to key processes leading the transition from oocyte to embryo, including meiotic spindle formation and positioning, regulation of translation, organelle redistribution, and epigenetic reprogramming. Yet, the underlying molecular mechanisms for these diverse functions are just beginning to be understood, hindered by unresolved interplay of SCMC components and variations in early lethal phenotypes. Here we review recent advances confirming involvement of the SCMC in human infertility, revealing an unexpected relationship with offspring health. Moreover, SCMC organization is being further revealed in terms of novel components and interactions with additional cell constituents. Collectively, this evidence prompts new avenues of investigation into possible roles during the process of oogenesis and the regulation of maternal transcript turnover during the oocyte to embryo transition.


Asunto(s)
Blastocisto/ultraestructura , Desarrollo Embrionario , Complejos Multiproteicos/fisiología , Oocitos/ultraestructura , Aneuploidia , Animales , Blastocisto/metabolismo , Anomalías Congénitas , Proteínas del Huevo/fisiología , Impresión Genómica , Humanos , Infertilidad/genética , Ratones , Complejos Multiproteicos/ultraestructura , Mutación , Oocitos/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo
5.
Life Sci Alliance ; 4(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34083394

RESUMEN

Nuclear organisation shapes gene regulation; however, the principles by which three-dimensional genome architecture influences gene transcription are incompletely understood. Condensin is a key architectural chromatin constituent, best known for its role in mitotic chromosome condensation. Yet at least a subset of condensin is bound to DNA throughout the cell cycle. Studies in various organisms have reported roles for condensin in transcriptional regulation, but no unifying mechanism has emerged. Here, we use rapid conditional condensin depletion in the budding yeast Saccharomyces cerevisiae to study its role in transcriptional regulation. We observe a large number of small gene expression changes, enriched at genes located close to condensin-binding sites, consistent with a possible local effect of condensin on gene expression. Furthermore, nascent RNA sequencing reveals that transcriptional down-regulation in response to environmental stimuli, in particular to heat shock, is subdued without condensin. Our results underscore the multitude by which an architectural chromosome constituent can affect gene regulation and suggest that condensin facilitates transcriptional reprogramming as part of adaptation to environmental changes.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , Expresión Génica/fisiología , Complejos Multiproteicos/metabolismo , Adenosina Trifosfatasas/fisiología , Cromatina/metabolismo , Segregación Cromosómica/fisiología , Cromosomas/metabolismo , ADN/fisiología , Proteínas de Unión al ADN/fisiología , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Mitosis/fisiología , Complejos Multiproteicos/fisiología , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
6.
Nat Struct Mol Biol ; 28(6): 501-511, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34117481

RESUMEN

The mammalian SWI/SNF complex, or BAF complex, has a conserved and direct role in antagonizing Polycomb-mediated repression. Yet, BAF also promotes repression by Polycomb in stem cells and cancer. How BAF both antagonizes and promotes Polycomb-mediated repression remains unknown. Here, we utilize targeted protein degradation to dissect the BAF-Polycomb axis in mouse embryonic stem cells on short timescales. We report that rapid BAF depletion redistributes Polycomb repressive complexes PRC1 and PRC2 from highly occupied domains, like Hox clusters, to weakly occupied sites normally opposed by BAF. Polycomb redistribution from highly repressed domains results in their decompaction, gain of active epigenomic features and transcriptional derepression. Surprisingly, through dose-dependent degradation of PRC1 and PRC2, we identify a conventional role for BAF in Polycomb-mediated repression, in addition to global Polycomb redistribution. These findings provide new mechanistic insight into the highly dynamic state of the Polycomb-Trithorax axis.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Represión Epigenética/fisiología , Regulación de la Expresión Génica/fisiología , Complejos Multiproteicos/fisiología , Proteínas del Grupo Polycomb/fisiología , Animales , Sistemas CRISPR-Cas , Células Cultivadas , Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/fisiología , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Represión Epigenética/genética , Edición Génica , Regulación de la Expresión Génica/genética , Genes Homeobox , Genoma , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Mutación con Pérdida de Función , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteolisis , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
8.
Science ; 372(6545): 984-989, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34045355

RESUMEN

We investigated genome folding across the eukaryotic tree of life. We find two types of three-dimensional (3D) genome architectures at the chromosome scale. Each type appears and disappears repeatedly during eukaryotic evolution. The type of genome architecture that an organism exhibits correlates with the absence of condensin II subunits. Moreover, condensin II depletion converts the architecture of the human genome to a state resembling that seen in organisms such as fungi or mosquitoes. In this state, centromeres cluster together at nucleoli, and heterochromatin domains merge. We propose a physical model in which lengthwise compaction of chromosomes by condensin II during mitosis determines chromosome-scale genome architecture, with effects that are retained during the subsequent interphase. This mechanism likely has been conserved since the last common ancestor of all eukaryotes.


Asunto(s)
Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/fisiología , Evolución Biológica , Cromosomas/ultraestructura , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Eucariontes/genética , Genoma , Complejos Multiproteicos/genética , Complejos Multiproteicos/fisiología , Adenosina Trifosfatasas/química , Algoritmos , Animales , Nucléolo Celular/ultraestructura , Núcleo Celular/ultraestructura , Centrómero/ultraestructura , Cromosomas/química , Cromosomas Humanos/química , Cromosomas Humanos/ultraestructura , Proteínas de Unión al ADN/química , Genoma Humano , Genómica , Heterocromatina/ultraestructura , Humanos , Interfase , Mitosis , Modelos Biológicos , Complejos Multiproteicos/química , Telómero/ultraestructura
9.
Mol Brain ; 14(1): 54, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33726789

RESUMEN

TMEM132D is a human gene identified with multiple risk alleles for panic disorders, anxiety and major depressive disorders. Defining a conserved family of transmembrane proteins, TMEM132D and its homologs are still of unknown molecular functions. By generating loss-of-function mutants of the sole TMEM132 ortholog in C. elegans, we identify abnormal morphologic phenotypes in the dopaminergic PDE neurons. Using a yeast two-hybrid screen, we find that NAP1 directly interacts with the cytoplasmic domain of human TMEM132D, and mutations in C. elegans tmem-132 that disrupt interaction with NAP1 cause similar morphologic defects in the PDE neurons. NAP1 is a component of the WAVE regulatory complex (WRC) that controls F-actin cytoskeletal dynamics. Decreasing activity of WRC rescues the PDE defects in tmem-132 mutants, whereas gain-of-function of TMEM132D in mammalian cells inhibits WRC, leading to decreased abundance of select WRC components, impaired actin nucleation and cell motility. We propose that metazoan TMEM132 family proteins play evolutionarily conserved roles in regulating NAP1 protein homologs to restrict inappropriate WRC activity, cytoskeletal and morphologic changes in the cell.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Citoesqueleto/ultraestructura , Neuronas Dopaminérgicas/ultraestructura , Proteínas de la Membrana/metabolismo , Morfogénesis/genética , Neurogénesis/genética , Células Receptoras Sensoriales/ultraestructura , Actinas/metabolismo , Animales , Evolución Biológica , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Forma de la Célula , Secuencia Conservada , Neuronas Dopaminérgicas/metabolismo , Mutación con Ganancia de Función , Genes Reporteros , Células HEK293 , Humanos , Mutación con Pérdida de Función , Familia de Multigenes , Complejos Multiproteicos/fisiología , Trastorno de Pánico/genética , Dominios Proteicos , Mapeo de Interacción de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Células Receptoras Sensoriales/metabolismo , Técnicas del Sistema de Dos Híbridos
10.
Nat Rev Mol Cell Biol ; 22(3): 196-213, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33510441

RESUMEN

Biomolecular condensates are membraneless intracellular assemblies that often form via liquid-liquid phase separation and have the ability to concentrate biopolymers. Research over the past 10 years has revealed that condensates play fundamental roles in cellular organization and physiology, and our understanding of the molecular principles, components and forces underlying their formation has substantially increased. Condensate assembly is tightly regulated in the intracellular environment, and failure to control condensate properties, formation and dissolution can lead to protein misfolding and aggregation, which are often the cause of ageing-associated diseases. In this Review, we describe the mechanisms and regulation of condensate assembly and dissolution, highlight recent advances in understanding the role of biomolecular condensates in ageing and disease, and discuss how cellular stress, ageing-related loss of homeostasis and a decline in protein quality control may contribute to the formation of aberrant, disease-causing condensates. Our improved understanding of condensate pathology provides a promising path for the treatment of protein aggregation diseases.


Asunto(s)
Envejecimiento , Sustancias Macromoleculares/química , Complejos Multiproteicos/fisiología , Agregación Patológica de Proteínas/etiología , Estrés Fisiológico/fisiología , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Fenómenos Fisiológicos Celulares , Humanos , Sustancias Macromoleculares/metabolismo , Agregado de Proteínas/fisiología , Agregación Patológica de Proteínas/metabolismo
11.
Nat Rev Mol Cell Biol ; 22(3): 183-195, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32632317

RESUMEN

Biomolecular condensation partitions cellular contents and has important roles in stress responses, maintaining homeostasis, development and disease. Many nuclear and cytoplasmic condensates are rich in RNA and RNA-binding proteins (RBPs), which undergo liquid-liquid phase separation (LLPS). Whereas the role of RBPs in condensates has been well studied, less attention has been paid to the contribution of RNA to LLPS. In this Review, we discuss the role of RNA in biomolecular condensation and highlight considerations for designing condensate reconstitution experiments. We focus on RNA properties such as composition, length, structure, modifications and expression level. These properties can modulate the biophysical features of native condensates, including their size, shape, viscosity, liquidity, surface tension and composition. We also discuss the role of RNA-protein condensates in development, disease and homeostasis, emphasizing how their properties and function can be determined by RNA. Finally, we discuss the multifaceted cellular functions of biomolecular condensates, including cell compartmentalization through RNA transport and localization, supporting catalytic processes, storage and inheritance of specific molecules, and buffering noise and responding to stress.


Asunto(s)
Sustancias Macromoleculares/química , Complejos Multiproteicos/química , Complejos Multiproteicos/fisiología , ARN/fisiología , Animales , Fenómenos Fisiológicos Celulares , Fenómenos Químicos , Humanos , Sustancias Macromoleculares/metabolismo , Complejos Multiproteicos/metabolismo , Agregado de Proteínas/fisiología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/fisiología
12.
Nucleic Acids Res ; 49(1): 206-220, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33330942

RESUMEN

Proteostasis needs to be tightly controlled to meet the cellular demand for correctly de novo folded proteins and to avoid protein aggregation. While a coupling between translation rate and co-translational folding, likely involving an interplay between the ribosome and its associated chaperones, clearly appears to exist, the underlying mechanisms and the contribution of ribosomal proteins remain to be explored. The ribosomal protein uL3 contains a long internal loop whose tip region is in close proximity to the ribosomal peptidyl transferase center. Intriguingly, the rpl3[W255C] allele, in which the residue making the closest contact to this catalytic site is mutated, affects diverse aspects of ribosome biogenesis and function. Here, we have uncovered, by performing a synthetic lethal screen with this allele, an unexpected link between translation and the folding of nascent proteins by the ribosome-associated Ssb-RAC chaperone system. Our results reveal that uL3 and Ssb-RAC cooperate to prevent 80S ribosomes from piling up within the 5' region of mRNAs early on during translation elongation. Together, our study provides compelling in vivo evidence for a functional connection between peptide bond formation at the peptidyl transferase center and chaperone-assisted de novo folding of nascent polypeptides at the solvent-side of the peptide exit tunnel.


Asunto(s)
Chaperonas Moleculares/fisiología , Complejos Multiproteicos/fisiología , Extensión de la Cadena Peptídica de Translación/fisiología , Pliegue de Proteína , Proteostasis/fisiología , Ribosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/metabolismo , Alelos , Mutación con Pérdida de Función , Chaperonas Moleculares/genética , Mutación Missense , Peptidil Transferasas/fisiología , Mutación Puntual , Proteínas Recombinantes/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/fisiología , Ribosomas/ultraestructura , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
13.
Nat Rev Mol Cell Biol ; 22(3): 215-235, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33169001

RESUMEN

Biomolecular condensates are found throughout eukaryotic cells, including in the nucleus, in the cytoplasm and on membranes. They are also implicated in a wide range of cellular functions, organizing molecules that act in processes ranging from RNA metabolism to signalling to gene regulation. Early work in the field focused on identifying condensates and understanding how their physical properties and regulation arise from molecular constituents. Recent years have brought a focus on understanding condensate functions. Studies have revealed functions that span different length scales: from molecular (modulating the rates of chemical reactions) to mesoscale (organizing large structures within cells) to cellular (facilitating localization of cellular materials and homeostatic responses). In this Roadmap, we discuss representative examples of biochemical and cellular functions of biomolecular condensates from the recent literature and organize these functions into a series of non-exclusive classes across the different length scales. We conclude with a discussion of areas of current interest and challenges in the field, and thoughts about how progress may be made to further our understanding of the widespread roles of condensates in cell biology.


Asunto(s)
Sustancias Macromoleculares , Complejos Multiproteicos/fisiología , Animales , Fenómenos Bioquímicos , Fenómenos Fisiológicos Celulares , Citoplasma/química , Citoplasma/genética , Citoplasma/metabolismo , Células Eucariotas/química , Células Eucariotas/metabolismo , Células Eucariotas/fisiología , Humanos , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Complejos Multiproteicos/química , Orgánulos/química , Orgánulos/genética , Orgánulos/metabolismo , Agregado de Proteínas/fisiología
14.
J Clin Endocrinol Metab ; 106(4): 1022-1040, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33382900

RESUMEN

CONTEXT: Low-grade chronic inflammation is commonly seen in polycystic ovary syndrome (PCOS) patients with elevated levels of inflammatory cytokines in the endometrium. OBJECTIVE: This work aimed to increase the limited understanding of the mechanisms underlying cytokine synthesis and increased endometrial inflammation in PCOS patients. METHODS: Endometrial biopsy samples were collected from non-PCOS (n = 17) and PCOS (n = 22) patients either during the proliferative phase of the menstrual cycle or with hyperplasia. Endometrial explants were prepared from PCOS patients and underwent pharmacological manipulation in vitro. The expression and localization of toll-like receptor 2 (TLR2)/4, key elements of innate immune signal transduction and nuclear factor κB (NFκB) signaling pathways, and multiple cytokines were comprehensively evaluated by Western blotting, immunohistochemistry, and immunofluorescence in endometrial tissues. RESULTS: We demonstrated the distribution of protein expression and localization associated with the significantly increased androgen receptor, TLR2, and TLR4-mediated activation of interferon regulatory factor-7 (IRF-7) and NFκB signaling, cytokine production, and endometrial inflammation in PCOS patients compared to non-PCOS patients with and without endometrial hyperplasia. In vitro experiments showed that 5-dihydrotestosterone (DHT) enhanced androgen receptor, TLR4, IRF-7, and p-NFκB p65 protein expression along with increased interferon α (IFNα) and IFNÉ£ abundance. The effects of DHT on IRF-7, p-NFκB p65, and IFN abundance were abolished by flutamide, an antiandrogen. Although 17ß-estradiol (E2) decreased p-IRF-7 expression with little effect on TLR-mediated IRF7 and NFκB signaling or on cytokine protein levels, exposure to metformin alone or in combination with E2 suppressed interleukin-1 receptor-associated kinase 4 (IRAK4), p-IRF-7, IRF-7, IκB kinase α (IKKα), p-NFκB p65, IFNÉ£, and tumor necrosis factor α protein expression. CONCLUSION: Cytokine synthesis and increased endometrial inflammation in PCOS patients are coupled to androgen-induced TLR4/IRF-7/NFκB signaling, which is inhibited by metformin treatment.


Asunto(s)
Andrógenos/farmacología , Endometrio/efectos de los fármacos , Metformina/farmacología , Síndrome del Ovario Poliquístico , Adulto , Citocinas/biosíntesis , Hiperplasia Endometrial/metabolismo , Hiperplasia Endometrial/patología , Endometrio/metabolismo , Endometrio/patología , Femenino , Humanos , Factor 7 Regulador del Interferón/metabolismo , Ciclo Menstrual/efectos de los fármacos , Ciclo Menstrual/metabolismo , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , FN-kappa B/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Unión Proteica , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo
15.
Blood ; 136(8): 974-988, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32584951

RESUMEN

Several adaptor molecules bind to cytoplasmic tails of ß-integrins and facilitate bidirectional signaling, which is critical in thrombosis and hemostasis. Interfering with integrin-adaptor interactions spatially or temporally to inhibit thrombosis without affecting hemostasis is an attractive strategy for the development of safe antithrombotic drugs. We show for the first time that the 14-3-3ζ-c-Src-integrin-ß3 complex is formed during platelet activation. 14-3-3ζ-c-Src interaction is mediated by the -PIRLGLALNFSVFYYE- fragment (PE16) on the 14-3-3ζ and SH2-domain on c-Src, whereas the 14-3-3ζ-integrin-ß3 interaction is mediated by the -ESKVFYLKMKGDYYRYL- fragment (EL17) on the 14-3-3ζ and -KEATSTF- fragment (KF7) on the ß3-integrin cytoplasmic tail. The EL17-motif inhibitor, or KF7 peptide, interferes with the formation of the 14-3-3ζ-c-Src-integrin-ß3 complex and selectively inhibits ß3 outside-in signaling without affecting the integrin-fibrinogen interaction, which suppresses thrombosis without causing significant bleeding. This study characterized a previously unidentified 14-3-3ζ-c-Src-integrin-ß3 complex in platelets and provided a novel strategy for the development of safe and effective antithrombotic treatments.


Asunto(s)
Proteínas 14-3-3/metabolismo , Integrina beta3/metabolismo , Activación Plaquetaria , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Proteínas 14-3-3/genética , Adulto , Animales , Femenino , Células HEK293 , Humanos , Integrina beta3/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , Activación Plaquetaria/genética , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Transducción de Señal/fisiología
16.
Sci Rep ; 10(1): 8321, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32433491

RESUMEN

Photoreceptors possess ribbon synapses distinct from the conventional synapses in the brain. Little is known about the function of the BBSome, a complex integral in ciliary and intracellular trafficking, in ribbon synaptic formation. We performed immunohistochemistry using retinas from Bardet-Biedl Syndrome (BBS) mouse models and found that BBS mutant animals have significantly fewer ribbon synapses in the outer plexiform layer and increased ectopic synapses in the outer nuclear layer compared to controls. Many ectopic synapses in BBS mutant retinas are associated with horizontal cell axonal processes that aberrantly intrude into the outer nuclear layer. To determine whether this horizontal cell phenotype is a consequence of retinal degeneration, we examined this phenotype in mice with photoreceptor-specific inactivation of the BBSome induced by Cre recombinase driven by the rhodopsin promoter. At three months of age, despite retinal degeneration, Bbs8floxed/floxed; Rho-Cre+ mice lack the aberrant intrusion of horizontal cell processes. At 6 months, some horizontal cell processes intrude into the outer nuclear layer in Bbs8floxed/floxed; Rho-Cre+ mice, but the phenotype does not recapitulate the phenotypic severity observed in young congenital BBS mutant mice. Therefore, the lack of BBSome function negatively impacts retinal synaptogenesis, and causes horizontal cell defects in a potentially cell-autonomous fashion.


Asunto(s)
Complejos Multiproteicos/fisiología , Células Fotorreceptoras/fisiología , Retina/patología , Sinapsis/patología , Animales , Síndrome de Bardet-Biedl , Cilios/fisiología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/fisiología , Femenino , Masculino , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Neurogénesis , Células Fotorreceptoras/patología , Proteínas/genética , Proteínas/fisiología , Degeneración Retiniana/patología
17.
Elife ; 92020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32391794

RESUMEN

Vesicle fusion is mediated by assembly of SNARE proteins between opposing membranes. While previous work suggested an active role of SNARE transmembrane domains (TMDs) in promoting membrane merger (Dhara et al., 2016), the underlying mechanism remained elusive. Here, we show that naturally-occurring v-SNARE TMD variants differentially regulate fusion pore dynamics in mouse chromaffin cells, indicating TMD flexibility as a mechanistic determinant that facilitates transmitter release from differentially-sized vesicles. Membrane curvature-promoting phospholipids like lysophosphatidylcholine or oleic acid profoundly alter pore expansion and fully rescue the decelerated fusion kinetics of TMD-rigidifying VAMP2 mutants. Thus, v-SNARE TMDs and phospholipids cooperate in supporting membrane curvature at the fusion pore neck. Oppositely, slowing of pore kinetics by the SNARE-regulator complexin-2 withstands the curvature-driven speeding of fusion, indicating that pore evolution is tightly coupled to progressive SNARE complex formation. Collectively, TMD-mediated support of membrane curvature and SNARE force-generated membrane bending promote fusion pore formation and expansion.


Asunto(s)
Exocitosis , Fusión de Membrana , Complejos Multiproteicos/fisiología , Neurotransmisores/fisiología , Fosfolípidos/metabolismo , Proteínas SNARE/fisiología , Proteína 2 de Membrana Asociada a Vesículas/fisiología , Animales , Calcio/fisiología , Membrana Celular/metabolismo , Células Cultivadas , Células Cromafines , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mutantes/fisiología , Unión Proteica , Dominios Proteicos , Vesículas Secretoras/fisiología
18.
Med Sci (Paris) ; 36(4): 367-375, 2020 Apr.
Artículo en Francés | MEDLINE | ID: mdl-32356713

RESUMEN

Human cytomegalovirus (HCMV) is an important ubiquitous opportunistic pathogen that belongs to the betaherpesviridae. Primary HCMV infection is generally asymptomatic in immunocompetent individuals. In contrast, HCMV infection causes serious disease in immunocompromised patients and is the leading cause of congenital viral infection. Although they are effective, the use of conventional molecules is limited by the emergence of resistance and by their toxicity. New antivirals targeting other replication steps and inducing fewer adverse effects are therefore needed. During HCMV replication, DNA packaging is performed by the terminase complex, which cleaves DNA to package the virus genome into the capsid. With no counterpart in mammalian cells, these terminase proteins are ideal targets for highly specific antivirals. A new terminase inhibitor, letermovir, recently proved effective against HCMV in phase III clinical trials. However, its mechanism of action is unclear and it has no significant activity against other herpesvirus or non-human CMV.


TITLE: Le complexe terminase, une cible de choix dans le traitement de l'infection à cytomégalovirus humain. ABSTRACT: Le cytomégalovirus humain (CMVH) est un pathogène opportuniste majeur en cas d'immunodépression et représente la principale cause d'infection congénitale d'origine virale. Bien qu'efficace, l'utilisation des molécules conventionnelles est limitée par leur toxicité et par l'émergence de résistance du virus, rendant nécessaire le développement de nouveaux traitements. Lors de la réplication du CMVH, l'encapsidation de l'ADN est réalisée par le complexe terminase qui clive l'ADN pour empaqueter le génome dans la capside. L'absence d'homologues dans les cellules des mammifères rend les protéines du complexe terminase des cibles idéales pour des antiviraux spécifiques. Une nouvelle molécule, le letermovir, cible une étape exclusivement virale en interagissant avec le complexe terminase. Son efficacité a été prouvée lors d'essais cliniques de phase III. Néanmoins, son mécanisme d'action n'est, à ce jour, pas élucidé et aucune activité n'est observée contre les autres herpèsvirus.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Endodesoxirribonucleasas/antagonistas & inhibidores , Terapia Molecular Dirigida/métodos , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/inmunología , Endodesoxirribonucleasas/fisiología , Humanos , Huésped Inmunocomprometido , Terapia Molecular Dirigida/tendencias , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/fisiología , Ensamble de Virus/efectos de los fármacos , Ensamble de Virus/fisiología , Replicación Viral/efectos de los fármacos
19.
Cell ; 181(3): 702-715.e20, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32315619

RESUMEN

Protein phosphatase 2A (PP2A) enzymes can suppress tumors, but they are often inactivated in human cancers overexpressing inhibitory proteins. Here, we identify a class of small-molecule iHAPs (improved heterocyclic activators of PP2A) that kill leukemia cells by allosterically assembling a specific heterotrimeric PP2A holoenzyme consisting of PPP2R1A (scaffold), PPP2R5E (B56ε, regulatory), and PPP2CA (catalytic) subunits. One compound, iHAP1, activates this complex but does not inhibit dopamine receptor D2, a mediator of neurologic toxicity induced by perphenazine and related neuroleptics. The PP2A complex activated by iHAP1 dephosphorylates the MYBL2 transcription factor on Ser241, causing irreversible arrest of leukemia and other cancer cells in prometaphase. In contrast, SMAPs, a separate class of compounds, activate PP2A holoenzymes containing a different regulatory subunit, do not dephosphorylate MYBL2, and arrest tumor cells in G1 phase. Our findings demonstrate that small molecules can serve as allosteric switches to activate distinct PP2A complexes with unique substrates.


Asunto(s)
Proteína Fosfatasa 2/metabolismo , Apoptosis , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Activadores de Enzimas/metabolismo , Fase G1 , Humanos , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , Fenotiazinas/farmacología , Fosforilación , Proteína Fosfatasa 2/fisiología , Subunidades de Proteína/metabolismo , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
20.
Blood ; 136(6): 684-697, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32325488

RESUMEN

The linear ubiquitin chain assembly complex (LUBAC) is a key regulator of NF-κB signaling. Activating single-nucleotide polymorphisms of HOIP, the catalytic subunit of LUBAC, are enriched in patients with activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL), and expression of HOIP, which parallels LUBAC activity, is elevated in ABC-DLBCL samples. Thus, to clarify the precise roles of LUBAC in lymphomagenesis, we generated a mouse model with augmented expression of HOIP in B cells. Interestingly, augmented HOIP expression facilitated DLBCL-like B-cell lymphomagenesis driven by MYD88-activating mutation. The developed lymphoma cells partly shared somatic gene mutations with human DLBCLs, with increased frequency of a typical AID mutation pattern. In vitro analysis revealed that HOIP overexpression protected B cells from DNA damage-induced cell death through NF-κB activation, and analysis of the human DLBCL database showed that expression of HOIP positively correlated with gene signatures representing regulation of apoptosis signaling, as well as NF-κB signaling. These results indicate that HOIP facilitates lymphomagenesis by preventing cell death and augmenting NF-κB signaling, leading to accumulation of AID-mediated mutations. Furthermore, a natural compound that specifically inhibits LUBAC was shown to suppress the tumor growth in a mouse transplantation model. Collectively, our data indicate that LUBAC is crucially involved in B-cell lymphomagenesis through protection against DNA damage-induced cell death and is a suitable therapeutic target for B-cell lymphomas.


Asunto(s)
Apoptosis/genética , Linfocitos B/enzimología , Transformación Celular Neoplásica/genética , Linfoma de Células B Grandes Difuso/etiología , Complejos Multiproteicos/fisiología , Ubiquitina-Proteína Ligasas/genética , Animales , Linfocitos B/patología , Proteínas Portadoras/fisiología , Daño del ADN , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Linfoma de Células B Grandes Difuso/enzimología , Linfoma de Células B Grandes Difuso/genética , Ratones , Ratones Transgénicos , Mutación Missense , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/fisiología , FN-kappa B/metabolismo , Trasplante de Neoplasias , Polimorfismo de Nucleótido Simple , Poliubiquitina/biosíntesis , Procesamiento Proteico-Postraduccional , Factores de Transcripción/fisiología , Transcriptoma , Ubiquitina-Proteína Ligasas/análisis , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación , Ubiquitinas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...