Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.152
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 185(20): 3739-3752.e18, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36113465

RESUMEN

Lysosomal amino acid efflux by proton-driven transporters is essential for lysosomal homeostasis, amino acid recycling, mTOR signaling, and maintaining lysosomal pH. To unravel the mechanisms of these transporters, we focus on cystinosin, a prototypical lysosomal amino acid transporter that exports cystine to the cytosol, where its reduction to cysteine supplies this limiting amino acid for diverse fundamental processes and controlling nutrient adaptation. Cystinosin mutations cause cystinosis, a devastating lysosomal storage disease. Here, we present structures of human cystinosin in lumen-open, cytosol-open, and cystine-bound states, which uncover the cystine recognition mechanism and capture the key conformational states of the transport cycle. Our structures, along with functional studies and double electron-electron resonance spectroscopic investigations, reveal the molecular basis for the transporter's conformational transitions and protonation switch, show conformation-dependent Ragulator-Rag complex engagement, and demonstrate an unexpected activation mechanism. These findings provide molecular insights into lysosomal amino acid efflux and a potential therapeutic strategy.


Asunto(s)
Cistina , Protones , Sistemas de Transporte de Aminoácidos/metabolismo , Cisteína/metabolismo , Cistina/metabolismo , Humanos , Lisosomas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
2.
Cell ; 180(6): 1115-1129.e13, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32200799

RESUMEN

Influenza A virus (IAV) is a lytic RNA virus that triggers receptor-interacting serine/threonine-protein kinase 3 (RIPK3)-mediated pathways of apoptosis and mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptosis in infected cells. ZBP1 initiates RIPK3-driven cell death by sensing IAV RNA and activating RIPK3. Here, we show that replicating IAV generates Z-RNAs, which activate ZBP1 in the nucleus of infected cells. ZBP1 then initiates RIPK3-mediated MLKL activation in the nucleus, resulting in nuclear envelope disruption, leakage of DNA into the cytosol, and eventual necroptosis. Cell death induced by nuclear MLKL was a potent activator of neutrophils, a cell type known to drive inflammatory pathology in virulent IAV disease. Consequently, MLKL-deficient mice manifest reduced nuclear disruption of lung epithelia, decreased neutrophil recruitment into infected lungs, and increased survival following a lethal dose of IAV. These results implicate Z-RNA as a new pathogen-associated molecular pattern and describe a ZBP1-initiated nucleus-to-plasma membrane "inside-out" death pathway with potentially pathogenic consequences in severe cases of influenza.


Asunto(s)
Virus de la Influenza A/genética , Necroptosis/genética , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis/genética , Muerte Celular/genética , Línea Celular Tumoral , Femenino , Virus de la Influenza A/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , ARN/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología
3.
Cell ; 179(5): 1057-1067.e14, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31730849

RESUMEN

The transition to a terrestrial environment, termed terrestrialization, is generally regarded as a pivotal event in the evolution and diversification of the land plant flora that changed the surface of our planet. Through phylogenomic studies, a group of streptophyte algae, the Zygnematophyceae, have recently been recognized as the likely sister group to land plants (embryophytes). Here, we report genome sequences and analyses of two early diverging Zygnematophyceae (Spirogloea muscicola gen. nov. and Mesotaenium endlicherianum) that share the same subaerial/terrestrial habitat with the earliest-diverging embryophytes, the bryophytes. We provide evidence that genes (i.e., GRAS and PYR/PYL/RCAR) that increase resistance to biotic and abiotic stresses in land plants, in particular desiccation, originated or expanded in the common ancestor of Zygnematophyceae and embryophytes, and were gained by horizontal gene transfer (HGT) from soil bacteria. These two Zygnematophyceae genomes represent a cornerstone for future studies to understand the underlying molecular mechanism and process of plant terrestrialization.


Asunto(s)
Evolución Biológica , Embryophyta/genética , Genoma de Planta , Streptophyta/genética , Ácido Abscísico/farmacología , Secuencia de Aminoácidos , Familia de Multigenes , Filogenia , Proteínas de Plantas/química , Dominios Proteicos , Streptophyta/clasificación , Simbiosis/genética , Sintenía/genética
4.
Cell ; 173(7): 1716-1727.e17, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29779945

RESUMEN

Sunlight exposure is known to affect mood, learning, and cognition. However, the molecular and cellular mechanisms remain elusive. Here, we show that moderate UV exposure elevated blood urocanic acid (UCA), which then crossed the blood-brain barrier. Single-cell mass spectrometry and isotopic labeling revealed a novel intra-neuronal metabolic pathway converting UCA to glutamate (GLU) after UV exposure. This UV-triggered GLU synthesis promoted its packaging into synaptic vesicles and its release at glutamatergic terminals in the motor cortex and hippocampus. Related behaviors, like rotarod learning and object recognition memory, were enhanced after UV exposure. All UV-induced metabolic, electrophysiological, and behavioral effects could be reproduced by the intravenous injection of UCA and diminished by the application of inhibitor or short hairpin RNA (shRNA) against urocanase, an enzyme critical for the conversion of UCA to GLU. These findings reveal a new GLU biosynthetic pathway, which could contribute to some of the sunlight-induced neurobehavioral changes.


Asunto(s)
Encéfalo/efectos de la radiación , Ácido Glutámico/biosíntesis , Aprendizaje/efectos de la radiación , Memoria/efectos de la radiación , Rayos Ultravioleta , Animales , Encéfalo/metabolismo , Encéfalo/patología , Cromatografía Líquida de Alta Presión , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/fisiología , Técnicas de Placa-Clamp , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Espectrometría de Masas en Tándem , Urocanato Hidratasa/antagonistas & inhibidores , Urocanato Hidratasa/genética , Urocanato Hidratasa/metabolismo , Ácido Urocánico/sangre , Ácido Urocánico/metabolismo
5.
Cell ; 173(3): 762-775.e16, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29677517

RESUMEN

Mechanotransduction plays a crucial role in vascular biology. One example of this is the local regulation of vascular resistance via flow-mediated dilation (FMD). Impairment of this process is a hallmark of endothelial dysfunction and a precursor to a wide array of vascular diseases, such as hypertension and atherosclerosis. Yet the molecules responsible for sensing flow (shear stress) within endothelial cells remain largely unknown. We designed a 384-well screening system that applies shear stress on cultured cells. We identified a mechanosensitive cell line that exhibits shear stress-activated calcium transients, screened a focused RNAi library, and identified GPR68 as necessary and sufficient for shear stress responses. GPR68 is expressed in endothelial cells of small-diameter (resistance) arteries. Importantly, Gpr68-deficient mice display markedly impaired acute FMD and chronic flow-mediated outward remodeling in mesenteric arterioles. Therefore, GPR68 is an essential flow sensor in arteriolar endothelium and is a critical signaling component in cardiovascular pathophysiology.


Asunto(s)
Mecanotransducción Celular , Interferencia de ARN , Receptores Acoplados a Proteínas G/fisiología , Animales , Materiales Biocompatibles , Calcio/metabolismo , Línea Celular Tumoral , Células Endoteliales/fisiología , Endotelio Vascular/citología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Concentración de Iones de Hidrógeno , Arterias Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/genética , Resistencia al Corte , Estrés Mecánico , Resistencia Vascular
6.
Nature ; 631(8020): 393-401, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38776962

RESUMEN

AMPylation is a post-translational modification in which AMP is added to the amino acid side chains of proteins1,2. Here we show that, with ATP as the ligand and actin as the host activator, the effector protein LnaB of Legionella pneumophila exhibits AMPylase activity towards the phosphoryl group of phosphoribose on PRR42-Ub that is generated by the SidE family of effectors, and deubiquitinases DupA and DupB in an E1- and E2-independent ubiquitination process3-7. The product of LnaB is further hydrolysed by an ADP-ribosylhydrolase, MavL, to Ub, thereby preventing the accumulation of PRR42-Ub and ADPRR42-Ub and protecting canonical ubiquitination in host cells. LnaB represents a large family of AMPylases that adopt a common structural fold, distinct from those of the previously known AMPylases, and LnaB homologues are found in more than 20 species of bacterial pathogens. Moreover, LnaB also exhibits robust phosphoryl AMPylase activity towards phosphorylated residues and produces unique ADPylation modifications in proteins. During infection, LnaB AMPylates the conserved phosphorylated tyrosine residues in the activation loop of the Src family of kinases8,9, which dampens downstream phosphorylation signalling in the host. Structural studies reveal the actin-dependent activation and catalytic mechanisms of the LnaB family of AMPylases. This study identifies, to our knowledge, an unprecedented molecular regulation mechanism in bacterial pathogenesis and protein phosphorylation.


Asunto(s)
Adenosina Monofosfato , Proteínas Bacterianas , Legionella pneumophila , Fosfotirosina , Transducción de Señal , Humanos , Actinas/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , ADP-Ribosilación , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Hidrólisis , Legionella pneumophila/enzimología , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidad , Ligandos , Modelos Moleculares , N-Glicosil Hidrolasas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Tirosina/metabolismo , Tirosina/química , Ubiquitina/metabolismo , Ubiquitinación , Enzimas Desubicuitinizantes/metabolismo , Pliegue de Proteína , Fosfotirosina/química , Fosfotirosina/metabolismo
7.
Immunity ; 52(2): 275-294.e9, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32075728

RESUMEN

Type 3 innate lymphoid cells (ILC3s) are critical for lung defense against bacterial pneumonia in the neonatal period, but the signals that guide pulmonary ILC3 development remain unclear. Here, we demonstrated that pulmonary ILC3s descended from ILC precursors that populated a niche defined by fibroblasts in the developing lung. Alveolar fibroblasts produced insulin-like growth factor 1 (IGF1), which instructed expansion and maturation of pulmonary ILC precursors. Conditional ablation of IGF1 in alveolar fibroblasts or deletion of the IGF-1 receptor from ILC precursors interrupted ILC3 biogenesis and rendered newborn mice susceptible to pneumonia. Premature infants with bronchopulmonary dysplasia, characterized by interrupted postnatal alveolar development and increased morbidity to respiratory infections, had reduced IGF1 concentrations and pulmonary ILC3 numbers. These findings indicate that the newborn period is a critical window in pulmonary immunity development, and disrupted lung development in prematurely born infants may have enduring effects on host resistance to respiratory infections.


Asunto(s)
Inmunidad Innata , Factor I del Crecimiento Similar a la Insulina/metabolismo , Pulmón/inmunología , Linfocitos/citología , Células Epiteliales Alveolares/metabolismo , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/inmunología , Diferenciación Celular , Proliferación Celular , Susceptibilidad a Enfermedades/inmunología , Humanos , Recién Nacido , Recien Nacido Prematuro , Factor I del Crecimiento Similar a la Insulina/deficiencia , Interleucinas/metabolismo , Pulmón/citología , Pulmón/crecimiento & desarrollo , Linfocitos/metabolismo , Ratones , Neumonía/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Interleucina-22
8.
Nature ; 620(7976): 1007-1012, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37364765

RESUMEN

Chemical transformations that introduce, remove or manipulate functional groups are ubiquitous in synthetic chemistry1. Unlike conventional functional-group interconversion reactions that swap one functionality for another, transformations that alter solely the location of functional groups are far less explored. Here, by photocatalytic, reversible C-H sampling, we report a functional-group translocation reaction of cyano (CN) groups in common nitriles, allowing for the direct positional exchange between a CN group and an unactivated C-H bond. The reaction shows high fidelity for 1,4-CN translocation, frequently contrary to inherent site selectivity in conventional C-H functionalizations. We also report the direct transannular CN translocation of cyclic systems, providing access to valuable structures that are non-trivial to obtain by other methods. Making use of the synthetic versatility of CN and a key CN translocation step, we showcase concise syntheses of building blocks of bioactive molecules. Furthermore, the combination of C-H cyanation and CN translocation allows access to unconventional C-H derivatives. Overall, the reported reaction represents a way to achieve site-selective C-H transformation reactions without requiring a site-selective C-H cleavage step.

9.
Cell ; 154(1): 103-17, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23827677

RESUMEN

Many neurodegenerative diseases are characterized by the accumulation of insoluble protein aggregates, including neurofibrillary tangles comprised of tau in Alzheimer's disease and Lewy bodies composed of α-synuclein in Parkinson's disease. Moreover, different pathological proteins frequently codeposit in disease brains. To test whether aggregated α-synuclein can directly cross-seed tau fibrillization, we administered preformed α-synuclein fibrils assembled from recombinant protein to primary neurons and transgenic mice. Remarkably, we discovered two distinct strains of synthetic α-synuclein fibrils that demonstrated striking differences in the efficiency of cross-seeding tau aggregation, both in neuron cultures and in vivo. Proteinase K digestion revealed conformational differences between the two synthetic α-synuclein strains and also between sarkosyl-insoluble α-synuclein extracted from two subgroups of Parkinson's disease brains. We speculate that distinct strains of pathological α-synuclein likely exist in neurodegenerative disease brains and may underlie the tremendous heterogeneity of synucleinopathies.


Asunto(s)
Neuronas/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Amiloide/química , Amiloide/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/metabolismo , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/química
10.
Nature ; 601(7892): 274-279, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34880492

RESUMEN

Glucose is a primary energy source in living cells. The discovery in 1960s that a sodium gradient powers the active uptake of glucose in the intestine1 heralded the concept of a secondary active transporter that can catalyse the movement of a substrate against an electrochemical gradient by harnessing energy from another coupled substrate. Subsequently, coupled Na+/glucose transport was found to be mediated by sodium-glucose cotransporters2,3 (SGLTs). SGLTs are responsible for active glucose and galactose absorption in the intestine and for glucose reabsorption in the kidney4, and are targeted by multiple drugs to treat diabetes5. Several members within the SGLT family transport key metabolites other than glucose2. Here we report cryo-electron microscopy structures of the prototypic human SGLT1 and a related monocarboxylate transporter SMCT1 from the same family. The structures, together with molecular dynamics simulations and functional studies, define the architecture of SGLTs, uncover the mechanism of substrate binding and selectivity, and shed light on water permeability of SGLT1. These results provide insights into the multifaceted functions of SGLTs.


Asunto(s)
Microscopía por Crioelectrón , Glucosa , Glucosa/metabolismo , Humanos , Transportadores de Ácidos Monocarboxílicos/química , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/ultraestructura , Sodio/metabolismo , Transportador 1 de Sodio-Glucosa/química , Transportador 1 de Sodio-Glucosa/metabolismo , Transportador 1 de Sodio-Glucosa/ultraestructura , Especificidad por Sustrato
11.
Nature ; 606(7914): 594-602, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35614224

RESUMEN

Only a small proportion of patients with cancer show lasting responses to immune checkpoint blockade (ICB)-based monotherapies. The RNA-editing enzyme ADAR1 is an emerging determinant of resistance to ICB therapy and prevents ICB responsiveness by repressing immunogenic double-stranded RNAs (dsRNAs), such as those arising from the dysregulated expression of endogenous retroviral elements (EREs)1-4. These dsRNAs trigger an interferon-dependent antitumour response by activating A-form dsRNA (A-RNA)-sensing proteins such as MDA-5 and PKR5. Here we show that ADAR1 also prevents the accrual of endogenous Z-form dsRNA elements (Z-RNAs), which were enriched in the 3' untranslated regions of interferon-stimulated mRNAs. Depletion or mutation of ADAR1 resulted in Z-RNA accumulation and activation of the Z-RNA sensor ZBP1, which culminated in RIPK3-mediated necroptosis. As no clinically viable ADAR1 inhibitors currently exist, we searched for a compound that can override the requirement for ADAR1 inhibition and directly activate ZBP1. We identified a small molecule, the curaxin CBL0137, which potently activates ZBP1 by triggering Z-DNA formation in cells. CBL0137 induced ZBP1-dependent necroptosis in cancer-associated fibroblasts and reversed ICB unresponsiveness in mouse models of melanoma. Collectively, these results demonstrate that ADAR1 represses endogenous Z-RNAs and identifies ZBP1-mediated necroptosis as a new determinant of tumour immunogenicity masked by ADAR1. Therapeutic activation of ZBP1-induced necroptosis provides a readily translatable avenue for rekindling the immune responsiveness of ICB-resistant human cancers.


Asunto(s)
Adenosina Desaminasa , Necroptosis , Neoplasias , Proteínas de Unión al ARN , Regiones no Traducidas 3' , Adenosina Desaminasa/metabolismo , Animales , Fibroblastos Asociados al Cáncer , Carbazoles/farmacología , Humanos , Inmunoterapia/tendencias , Interferones/metabolismo , Melanoma , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , ARN Bicatenario/inmunología , Proteínas de Unión al ARN/metabolismo
12.
Nature ; 596(7872): 444-448, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34349262

RESUMEN

MFSD2A is a sodium-dependent lysophosphatidylcholine symporter that is responsible for the uptake of docosahexaenoic acid into the brain1,2, which is crucial for the development and performance of the brain3. Mutations that affect MFSD2A cause microcephaly syndromes4,5. The ability of MFSD2A to transport lipid is also a key mechanism that underlies its function as an inhibitor of transcytosis to regulate the blood-brain barrier6,7. Thus, MFSD2A represents an attractive target for modulating the permeability of the blood-brain barrier for drug delivery. Here we report the cryo-electron microscopy structure of mouse MFSD2A. Our structure defines the architecture of this important transporter, reveals its unique extracellular domain and uncovers its substrate-binding cavity. The structure-together with our functional studies and molecular dynamics simulations-identifies a conserved sodium-binding site, reveals a potential lipid entry pathway and helps to rationalize MFSD2A mutations that underlie microcephaly syndromes. These results shed light on the critical lipid transport function of MFSD2A and provide a framework to aid in the design of specific modulators for therapeutic purposes.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Metabolismo de los Lípidos , Simportadores/química , Simportadores/metabolismo , Animales , Sitios de Unión , Transporte Biológico , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación , Dominios Proteicos , Sodio/metabolismo , Simportadores/genética , Simportadores/ultraestructura
13.
Physiol Rev ; 99(1): 513-554, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30427276

RESUMEN

The unique architecture of the mammalian lung is required for adaptation to air breathing at birth and thereafter. Understanding the cellular and molecular mechanisms controlling its morphogenesis provides the framework for understanding the pathogenesis of acute and chronic lung diseases. Recent single-cell RNA sequencing data and high-resolution imaging identify the remarkable heterogeneity of pulmonary cell types and provides cell selective gene expression underlying lung development. We will address fundamental issues related to the diversity of pulmonary cells, to the formation and function of the mammalian lung, and will review recent advances regarding the cellular and molecular pathways involved in lung organogenesis. What cells form the lung in the early embryo? How are cell proliferation, migration, and differentiation regulated during lung morphogenesis? How do cells interact during lung formation and repair? How do signaling and transcriptional programs determine cell-cell interactions necessary for lung morphogenesis and function?


Asunto(s)
Diferenciación Celular/fisiología , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Pulmón/citología , Morfogénesis/fisiología , Animales , Proliferación Celular/fisiología , Desarrollo Embrionario/genética , Humanos , Pulmón/metabolismo , Morfogénesis/genética
14.
Hum Mol Genet ; 33(12): 1064-1073, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38507061

RESUMEN

Cancer-associated fibroblasts (CAFs) are increasingly recognized as playing a crucial role in regulating cancer progression and metastasis. These cells can be activated by long non-coding RNAs (lncRNAs), promoting the malignant biological processes of tumor cells. Therefore, it is essential to understand the regulatory relationship between CAFs and lncRNAs in cancers. Here, we identified CAF-related lncRNAs at the pan-cancer level to systematically predict their potential regulatory functions. The identified lncRNAs were also validated using various external data at both tissue and cellular levels. This study has revealed that these CAF-related lncRNAs exhibit expression perturbations in cancers and are highly correlated with the infiltration of stromal cells, particularly fibroblasts and endothelial cells. By prioritizing a list of CAF-related lncRNAs, we can further distinguish patient subtypes that show survival and molecular differences. In addition, we have developed a web server, CAFLnc (https://46906u5t63.zicp.fun/CAFLnc/), to visualize our results. In conclusion, CAF-related lncRNAs hold great potential as a valuable resource for comprehending lncRNA functions and advancing the identification of biomarkers for cancer progression and therapeutic targets in cancer treatment.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinogénesis , Regulación Neoplásica de la Expresión Génica , Neoplasias , ARN Largo no Codificante , ARN Largo no Codificante/genética , Humanos , Neoplasias/genética , Neoplasias/patología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinogénesis/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Perfilación de la Expresión Génica , Microambiente Tumoral/genética
15.
EMBO J ; 41(17): e108368, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35801308

RESUMEN

The evolutionary benefit accounting for widespread conservation of oligomeric structures in proteins lacking evidence of intersubunit cooperativity remains unclear. Here, crystal and cryo-EM structures, and enzymological data, demonstrate that a conserved tetramer interface maintains the active-site structure in one such class of proteins, the short-chain dehydrogenase/reductase (SDR) superfamily. Phylogenetic comparisons support a significantly longer polypeptide being required to maintain an equivalent active-site structure in the context of a single subunit. Oligomerization therefore enhances evolutionary fitness by reducing the metabolic cost of enzyme biosynthesis. The large surface area of the structure-stabilizing oligomeric interface yields a synergistic gain in fitness by increasing tolerance to activity-enhancing yet destabilizing mutations. We demonstrate that two paralogous SDR superfamily enzymes with different specificities can form mixed heterotetramers that combine their individual enzymological properties. This suggests that oligomerization can also diversify the functions generated by a given metabolic investment, enhancing the fitness advantage provided by this architectural strategy.


Asunto(s)
Evolución Biológica , Oxidorreductasas , Secuencia de Aminoácidos , Dominio Catalítico , Oxidorreductasas/metabolismo , Filogenia
16.
EMBO J ; 41(11): e109902, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35343600

RESUMEN

Induction and suppression of antiviral RNA interference (RNAi) has been observed in mammals during infection with at least seven distinct RNA viruses, including some that are pathogenic in humans. However, while the cell-autonomous immune response mediated by antiviral RNAi is gradually being recognized, little is known about systemic antiviral RNAi in mammals. Furthermore, extracellular vesicles (EVs) also function in viral signal spreading and host immunity. Here, we show that upon antiviral RNAi activation, virus-derived small-interfering RNAs (vsiRNAs) from Nodamura virus (NoV), Sindbis virus (SINV), and Zika virus (ZIKV) enter the murine bloodstream via EVs for systemic circulation. vsiRNAs in the EVs are biologically active, since they confer RNA-RNA homology-dependent antiviral activity in both cultured cells and infant mice. Moreover, we demonstrate that vaccination with a live-attenuated virus, rendered deficient in RNAi suppression, induces production of stably maintained vsiRNAs and confers protective immunity against virus infection in mice. This suggests that vaccination with live-attenuated VSR (viral suppressor of RNAi)-deficient mutant viruses could be a new strategy to induce immunity.


Asunto(s)
Vesículas Extracelulares , Infección por el Virus Zika , Virus Zika , Animales , Antivirales , Vesículas Extracelulares/genética , Humanos , Mamíferos/genética , Ratones , Interferencia de ARN , ARN Bicatenario , ARN Interferente Pequeño/genética , Virus Zika/genética , Infección por el Virus Zika/genética , Infección por el Virus Zika/prevención & control
17.
Nat Chem Biol ; 20(4): 463-472, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37945894

RESUMEN

Ubiquitination plays essential roles in eukaryotic cellular processes. The effector protein CteC from Chromobacterium violaceum blocks host ubiquitination by mono-ADP-ribosylation of ubiquitin (Ub) at residue T66. However, the structural basis for this modification is unknown. Here we report three crystal structures of CteC in complexes with Ub, NAD+ or ADP-ribosylated Ub, which represent different catalytic states of CteC in the modification. CteC adopts a special 'D-E' catalytic motif for catalysis and binds NAD+ in a half-ligand binding mode. The specific recognition of Ub by CteC is determined by a relatively separate Ub-targeting domain and a long loop L6, not the classic ADP-ribosylating turn-turn loop. Structural analyses with biochemical results reveal that CteC represents a large family of poly (ADP-ribose) polymerase (PARP)-like ADP-ribosyltransferases, which harbors chimeric features from the R-S-E and H-Y-E classes of ADP-ribosyltransferases. The family of CteC-like ADP-ribosyltransferases has a common 'D-E' catalytic consensus and exists extensively in bacteria and eukaryotic microorganisms.


Asunto(s)
Treonina , Ubiquitina , Ubiquitina/química , Treonina/metabolismo , NAD/metabolismo , ADP-Ribosilación , ADP Ribosa Transferasas/química , Poli(ADP-Ribosa) Polimerasas/química , Bacterias/metabolismo , Adenosina Difosfato Ribosa
18.
Nature ; 582(7810): 129-133, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32494073

RESUMEN

Mitochondria take up Ca2+ through the mitochondrial calcium uniporter complex to regulate energy production, cytosolic Ca2+ signalling and cell death1,2. In mammals, the uniporter complex (uniplex) contains four core components: the pore-forming MCU protein, the gatekeepers MICU1 and MICU2, and an auxiliary subunit, EMRE, essential for Ca2+ transport3-8. To prevent detrimental Ca2+ overload, the activity of MCU must be tightly regulated by MICUs, which sense changes in cytosolic Ca2+ concentrations to switch MCU on and off9,10. Here we report cryo-electron microscopic structures of the human mitochondrial calcium uniporter holocomplex in inhibited and Ca2+-activated states. These structures define the architecture of this multicomponent Ca2+-uptake machinery and reveal the gating mechanism by which MICUs control uniporter activity. Our work provides a framework for understanding regulated Ca2+ uptake in mitochondria, and could suggest ways of modulating uniporter activity to treat diseases related to mitochondrial Ca2+ overload.


Asunto(s)
Canales de Calcio/química , Canales de Calcio/metabolismo , Microscopía por Crioelectrón , Sitios de Unión/efectos de los fármacos , Calcio/metabolismo , Calcio/farmacología , Canales de Calcio/ultraestructura , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura
19.
Proc Natl Acad Sci U S A ; 120(5): e2214684120, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36693099

RESUMEN

Embryo implantation, a crucial step in human reproduction, is tightly controlled by estrogen and progesterone (P4) via estrogen receptor alpha and progesterone receptor (PGR), respectively. Here, we report that N6-methyladenosine (m6A), the most abundant mRNA modification in eukaryotes, plays an essential role in embryo implantation through the maintenance of P4 signaling. Conditional deletion of methyltransferase-like 3 (Mettl3), encoding the m6A writer METTL3, in the female reproductive tract using a Cre mouse line with Pgr promoter (Pgr-Cre) resulted in complete implantation failure due to pre-implantation embryo loss and defective uterine receptivity. Moreover, the uterus of Mettl3 null mice failed to respond to artificial decidualization. We further found that Mettl3 deletion was accompanied by a marked decrease in PGR protein expression. Mechanistically, we found that Pgr mRNA is a direct target for METTL3-mediated m6A modification. A luciferase assay revealed that the m6A modification in the 5' untranslated region (5'-UTR) of Pgr mRNA enhances PGR protein translation efficiency in a YTHDF1-dependent manner. Finally, we demonstrated that METTL3 is required for human endometrial stromal cell decidualization in vitro and that the METTL3-PGR axis is conserved between mice and humans. In summary, this study provides evidence that METTL3 is essential for normal P4 signaling during embryo implantation via m6A-mediated translation control of Pgr mRNA.


Asunto(s)
Progesterona , Receptores de Progesterona , Femenino , Ratones , Humanos , Animales , Progesterona/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Implantación del Embrión/genética , Útero/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones Noqueados , ARN Mensajero/metabolismo
20.
Plant J ; 118(2): 324-344, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38149487

RESUMEN

Sclerotinia sclerotiorum causes white mold or stem rot in a wide range of economically important plants, bringing significant yield losses worldwide. Control of this pathogen is difficult as its resting structure sclerotia can survive in soil for years, and no Resistance genes have been identified in S. sclerotiorum hosts. Host-induced gene silencing (HIGS) has shown promising effects in controlling many fungal pathogens, including S. sclerotiorum. However, better molecular genetic understanding of signaling pathways involved in its development and pathogenicity is needed to provide effective HIGS gene targets. Here, by employing a forward genetic screen, we characterized an evolutionarily conserved mitogen-activated protein kinase (MAPK) cascade in S. sclerotiorum, consisting of SsSte50-SsSte11-SsSte7-Smk1, which controls mycelial growth, sclerotia development, compound appressoria formation, virulence, and hyphal fusion. Moreover, disruption of the putative downstream transcription factor SsSte12 led to normal sclerotia but deformed appressoria and attenuated host penetration, as well as impaired apothecia formation, suggestive of diverged regulation downstream of the MAPK cascade. Most importantly, targeting SsSte50 using host-expressed double-stranded RNA resulted in largely reduced virulence of S. sclerotiorum on both Nicotiana benthamiana leaves and transgenic Arabidopsis thaliana plants. Therefore, this MAPK signaling cascade is generally needed for its growth, development, and pathogenesis and can serve as ideal HIGS targets for mitigating economic damages caused by S. sclerotiorum infection.


Asunto(s)
Ascomicetos , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Hifa , Silenciador del Gen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA