Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.940
Filtrar
1.
Methods Mol Biol ; 2799: 29-46, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38727901

RESUMEN

The expression and activity of ionotropic glutamate receptors control signal transduction at the excitatory synapses in the CNS. The NMDAR comprises two obligatory GluN1 subunits and two GluN2 or GluN3 subunits in different combinations. Each GluN subunit consists of four domains: the extracellular amino-terminal and agonist-binding domains, the transmembrane domain, and the intracellular C-terminal domain (CTD). The CTD interaction with various classes of intracellular proteins is critical for trafficking and synaptic localization of NMDARs. Amino acid mutations or the inclusion of premature stop codons in the CTD could contribute to the emergence of neurodevelopmental and neuropsychiatric disorders. Here, we describe the method of preparing primary hippocampal neurons and lentiviral particles expressing GluN subunits that can be used as a model to study cell surface expression and synaptic localization of NMDARs. We also show a simple method of fluorescence immunostaining of eGFP-tagged GluN2 subunits and subsequent microscopy technique and image analysis to study the effects of disease-associated mutations in the CTDs of GluN2A and GluN2B subunits.


Asunto(s)
Hipocampo , Neuronas , Receptores de N-Metil-D-Aspartato , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Hipocampo/metabolismo , Hipocampo/citología , Neuronas/metabolismo , Animales , Subunidades de Proteína/metabolismo , Subunidades de Proteína/genética , Células Cultivadas , Ratas , Humanos , Lentivirus/genética , Cultivo Primario de Células/métodos , Expresión Génica
2.
Methods Mol Biol ; 2799: 139-150, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38727906

RESUMEN

Epilepsy is one of the most represented neurological diseases worldwide. However, in many cases, the precise molecular mechanisms of epileptogenesis and ictiogenesis are unknown. Because of their important role in synaptic function and neuronal excitability, NMDA receptors are implicated in various epileptogenic mechanisms. Most of these are subunit specific and require a precise analysis of the subunit composition of the NMDARs implicated. Here, we describe an express electrophysiological method to analyze the contribution of NMDAR subunits to spontaneous postsynaptic activity in identified cells in brain slices using patch clamp whole cell recordings.


Asunto(s)
Técnicas de Placa-Clamp , Receptores de N-Metil-D-Aspartato , Sinapsis , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Técnicas de Placa-Clamp/métodos , Sinapsis/metabolismo , Sinapsis/fisiología , Encéfalo/metabolismo , Encéfalo/citología , Neuronas/metabolismo , Ratones , Ratas , Subunidades de Proteína/metabolismo
3.
Methods Mol Biol ; 2799: 55-77, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38727903

RESUMEN

NMDA-type ionotropic glutamate receptors are critically involved in many brain functions and are implicated in a variety of brain disorders. Seven NMDA receptor subunits exist (GluN1, GluN2A-D, and GluN3A-B) that assemble into tetrameric receptor subtypes with distinct functional properties and physiological roles. The majority NMDA receptors are composed of two GluN1 and two GluN2 subunits, which can assemble into four diheteromeric receptors subtypes composed of GluN1 and one type of GluN2 subunit (e.g., GluN1/2A), and presumably also six triheteromeric receptor subtypes composed of GluN1 and two different GluN2 subunits (e.g., GluN1/2A/2B). Furthermore, the GluN1 subunit exists as eight splice variants (e.g., GluN1-1a and GluN1-1b isoforms), and two different GluN1 isoforms can co-assemble to also form triheteromeric NMDA receptors (e.g., GluN1-1a/1b/2A). Here, we describe a method to faithfully express triheteromeric NMDA receptors in heterologous expression systems by controlling the identity of two of the four subunits. This method overcomes the problem that co-expression of three different NMDA receptor subunits generates two distinct diheteromeric receptor subtypes as well as one triheteromeric receptor subtype, thereby confounding studies that require a homogenous population of triheteromeric NMDA receptors. The method has been applied to selectively express recombinant triheteromeric GluN1/2A/2B, GluN1/2A/2C, GluN1/2B/2D, GluN1-1a/GluN1-1b/2A, GluN1-1a/GluN1-1b/2B receptors with negligible co-expression of the respective diheteromeric receptor subtypes. This method therefore enables quantitative evaluation of functional and pharmacological properties of triheteromeric NMDA receptors, some of which are abundant NMDA receptor subtypes in the adult brain.


Asunto(s)
Isoformas de Proteínas , Subunidades de Proteína , Receptores de N-Metil-D-Aspartato , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Humanos , Subunidades de Proteína/metabolismo , Subunidades de Proteína/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Células HEK293 , Animales , Membrana Celular/metabolismo , Expresión Génica
4.
Methods Mol Biol ; 2799: 257-267, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38727912

RESUMEN

The NMDAR is a heterotetramer composed of two GluN1 subunits and two GluN2 and/or GluN3 subunits, with the GluN2 subunits exhibiting significant diversity in their structure and function. Recent studies have highlighted the importance of characterizing the specific roles of each GluN2 subunit across central nervous system regions and developmental stages, as well as their unique contributions to NMDAR-mediated signaling and plasticity. Understanding the distinct functions of GluN2 subunits is critical for the development of targeted therapeutic strategies for NMDAR-related disorders. However, measuring the functional contribution of individual GluN2 subtypes in ex vivo slices is challenging. Conventionally, pharmacological or genetic approaches are used, but, in many cases, this is not possible or is restricted to population-level NMDAR responses. Here, we describe a technique for using biophysical properties of miniature synaptic NMDAR responses as a proxy to measure the functional contribution of specific GluN2-NMDAR subunits to individual synapses within a neuron.


Asunto(s)
Subunidades de Proteína , Receptores de N-Metil-D-Aspartato , Sinapsis , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Animales , Sinapsis/metabolismo , Subunidades de Proteína/metabolismo , Ratones , Neuronas/metabolismo , Ratas , Técnicas de Placa-Clamp/métodos , Transmisión Sináptica
5.
Sci Rep ; 14(1): 10146, 2024 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-38698024

RESUMEN

The closely related endolysosomal tethering complexes HOPS and CORVET play pivotal roles in the homo- and heterotypic fusion of early and late endosomes, respectively, and HOPS also mediates the fusion of lysosomes with incoming vesicles including late endosomes and autophagosomes. These heterohexameric complexes share their four core subunits that assemble with additional two, complex-specific subunits. These features and the similar structure of the complexes could allow the formation of hybrid complexes, and the complex specific subunits may compete for binding to the core. Indeed, our biochemical analyses revealed the overlap of binding sites for HOPS-specific VPS41 and CORVET-specific VPS8 on the shared core subunit VPS18. We found that the overexpression of CORVET-specific VPS8 or Tgfbrap1 decreased the amount of core proteins VPS11 and VPS18 that are assembled with HOPS-specific subunits VPS41 or VPS39, indicating reduced amount of assembled HOPS. In line with this, we observed the elevation of both lipidated, autophagosome-associated LC3 protein and the autophagic cargo p62 in these cells, suggesting impaired autophagosome-lysosome fusion. In contrast, overexpression of HOPS-specific VPS39 or VPS41 did not affect the level of assembled CORVET or autophagy. VPS8 or Tgfbrap1 overexpression also induced Cathepsin D accumulation, suggesting that HOPS-dependent biosynthetic delivery of lysosomal hydrolases is perturbed, too. These indicate that CORVET-specific subunit levels fine-tune HOPS assembly and activity in vivo.


Asunto(s)
Endosomas , Proteínas de Transporte Vesicular , Endosomas/metabolismo , Humanos , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/genética , Lisosomas/metabolismo , Subunidades de Proteína/metabolismo , Autofagia , Autofagosomas/metabolismo , Células HeLa , Unión Proteica
6.
Bioorg Med Chem ; 106: 117733, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38704960

RESUMEN

Development of selective or dual proteasome subunit inhibitors based on syringolin B as a scaffold is described. We focused our efforts on a structure-activity relationship study of inhibitors with various substituents at the 3-position of the macrolactam moiety of syringolin B analogue to evaluate whether this would be sufficient to confer subunit selectivity by using sets of analogues with hydrophobic, basic and acidic substituents, which were designed to target Met45, Glu53 and Arg45 embedded in the S1 subsite, respectively. The structure-activity relationship study using systematic analogues provided insight into the origin of the subunit-selective inhibitory activity. This strategy would be sufficient to confer subunit selectivity regarding ß5 and ß2 subunits.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Inhibidores de Proteasoma , Relación Estructura-Actividad , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/síntesis química , Humanos , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Subunidades de Proteína/química , Estructura Molecular
7.
Biochem Biophys Res Commun ; 718: 150087, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38735139

RESUMEN

Flooding deprives plants of oxygen and thereby causes severe stress by interfering with energy production, leading to growth retardation. Enzymes and metabolites may help protect plants from waterlogging and hypoxic environmental conditions. Acetolactate synthase (ALS) is a key enzyme in the biosynthesis of branched-chain amino acids (BCAAs), providing the building blocks for proteins and various secondary metabolites. Additionally, under energy-poor conditions, free BCAAs can be used as an alternative energy source by mitochondria through a catabolic enzyme chain reaction. In this study, we characterized ALS-INTERACTING PROTEIN 1 (OsAIP1), which encodes the regulatory subunit of ALS in rice (Oryza sativa). This gene was expressed in all parts of the rice plant, and its expression level was significantly higher in submerged and low-oxygen environments. Rice transformants overexpressing OsAIP1 showed a higher survival rate under hypoxic stress than did non-transgenic control plants under the same conditions. The OsAIP1-overexpressing plants accumulated increased levels of BCAAs, demonstrating that OsAIP1 is an important factor in the hypoxia resistance mechanism. These results suggest that ALS proteins are part of a defense mechanism that improves the tolerance of plants to low-oxygen environments.


Asunto(s)
Acetolactato Sintasa , Regulación de la Expresión Génica de las Plantas , Oryza , Proteínas de Plantas , Oryza/genética , Oryza/metabolismo , Oryza/enzimología , Acetolactato Sintasa/genética , Acetolactato Sintasa/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente , Estrés Fisiológico/genética , Aminoácidos de Cadena Ramificada/metabolismo , Oxígeno/metabolismo , Subunidades de Proteína/metabolismo , Subunidades de Proteína/genética
8.
Int J Mol Sci ; 25(9)2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38732022

RESUMEN

The molecular weight (MW) of an enzyme is a critical parameter in enzyme-constrained models (ecModels). It is determined by two factors: the presence of subunits and the abundance of each subunit. Although the number of subunits (NS) can potentially be obtained from UniProt, this information is not readily available for most proteins. In this study, we addressed this gap by extracting and curating subunit information from the UniProt database to establish a robust benchmark dataset. Subsequently, we propose a novel model named DeepSub, which leverages the protein language model and Bi-directional Gated Recurrent Unit (GRU), to predict NS in homo-oligomers solely based on protein sequences. DeepSub demonstrates remarkable accuracy, achieving an accuracy rate as high as 0.967, surpassing the performance of QUEEN. To validate the effectiveness of DeepSub, we performed predictions for protein homo-oligomers that have been reported in the literature but are not documented in the UniProt database. Examples include homoserine dehydrogenase from Corynebacterium glutamicum, Matrilin-4 from Mus musculus and Homo sapiens, and the Multimerins protein family from M. musculus and H. sapiens. The predicted results align closely with the reported findings in the literature, underscoring the reliability and utility of DeepSub.


Asunto(s)
Bases de Datos de Proteínas , Aprendizaje Profundo , Subunidades de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Animales , Humanos , Multimerización de Proteína , Ratones , Biología Computacional/métodos
9.
J Mol Biol ; 436(10): 168568, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38583515

RESUMEN

Porphyromonas gingivalis, an anaerobic CFB (Cytophaga, Fusobacterium, and Bacteroides) group bacterium, is the keystone pathogen of periodontitis and has been implicated in various systemic diseases. Increased antibiotic resistance and lack of effective antibiotics necessitate a search for new intervention strategies. Here we report a 3.5 Å resolution cryo-EM structure of P. gingivalis RNA polymerase (RNAP). The structure displays new structural features in its ω subunit and multiple domains in ß and ß' subunits, which differ from their counterparts in other bacterial RNAPs. Superimpositions with E. coli RNAP holoenzyme and initiation complex further suggest that its ω subunit may contact the σ4 domain, thereby possibly contributing to the assembly and stabilization of initiation complexes. In addition to revealing the unique features of P. gingivalis RNAP, our work offers a framework for future studies of transcription regulation in this important pathogen, as well as for structure-based drug development.


Asunto(s)
Microscopía por Crioelectrón , ARN Polimerasas Dirigidas por ADN , Modelos Moleculares , Porphyromonas gingivalis , Porphyromonas gingivalis/enzimología , Porphyromonas gingivalis/genética , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/ultraestructura , ARN Polimerasas Dirigidas por ADN/genética , Conformación Proteica , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/enzimología , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo
10.
Photosynth Res ; 160(2-3): 87-96, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38625595

RESUMEN

The primary photochemical reaction of photosynthesis in green sulfur bacteria occurs in the homodimer PscA core proteins by a special chlorophyll pair. The light induced excited state of the special pair producing P840+ is rapidly reduced by electron transfer from one of the two PscC subunits. Molecular dynamics (MD) simulations are combined with bioinformatic tools herein to provide structural and dynamic insight into the complex between the two PscA core proteins and the two PscC subunits. The microscopic dynamic model involves extensive sampling at atomic resolution and at a cumulative time-scale of 22µs and reveals well defined protein-protein interactions. The membrane complex is composed of the two PscA and the two PscC subunits and macroscopic connections are revealed within a putative electron transfer pathway from the PscC subunit to the special pair P840 located within the PscA subunits. Our results provide a structural basis for understanding the electron transport to the homodimer RC of the green sulfur bacteria. The MD based approach can provide the basis to further probe the PscA-PscC complex dynamics and observe electron transfer therein at the quantum level. Furthermore, the transmembrane helices of the different PscC subunits exert distinct dynamics in the complex.


Asunto(s)
Proteínas Bacterianas , Chlorobi , Simulación de Dinámica Molecular , Transporte de Electrón , Chlorobi/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Subunidades de Proteína/metabolismo , Subunidades de Proteína/química , Fotosíntesis , Clorofila/metabolismo , Complejos de Proteína Captadores de Luz/metabolismo , Complejos de Proteína Captadores de Luz/química
11.
Cell Rep ; 43(4): 114011, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38573854

RESUMEN

Fatalska et al.1 use an interdisciplinary strategy to elucidate how an intrinsically disordered regulatory subunit of protein phosphatase 1 binds trimeric eIF2 and positions the phosphatase-substrate complex for dephosphorylation. As validation, they show that a disease mutation abolishes the interaction.


Asunto(s)
Factor 2 Eucariótico de Iniciación , Proteína Fosfatasa 1 , Proteína Fosfatasa 1/metabolismo , Humanos , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Unión Proteica , Fosforilación , Subunidades de Proteína/metabolismo , Subunidades de Proteína/química , Mutación
12.
Nature ; 629(8010): 219-227, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38570683

RESUMEN

The Integrator complex can terminate RNA polymerase II (Pol II) in the promoter-proximal region of genes. Previous work has shed light on how Integrator binds to the paused elongation complex consisting of Pol II, the DRB sensitivity-inducing factor (DSIF) and the negative elongation factor (NELF) and how it cleaves the nascent RNA transcript1, but has not explained how Integrator removes Pol II from the DNA template. Here we present three cryo-electron microscopy structures of the complete Integrator-PP2A complex in different functional states. The structure of the pre-termination complex reveals a previously unresolved, scorpion-tail-shaped INTS10-INTS13-INTS14-INTS15 module that may use its 'sting' to open the DSIF DNA clamp and facilitate termination. The structure of the post-termination complex shows that the previously unresolved subunit INTS3 and associated sensor of single-stranded DNA complex (SOSS) factors prevent Pol II rebinding to Integrator after termination. The structure of the free Integrator-PP2A complex in an inactive closed conformation2 reveals that INTS6 blocks the PP2A phosphatase active site. These results lead to a model for how Integrator terminates Pol II transcription in three steps that involve major rearrangements.


Asunto(s)
Microscopía por Crioelectrón , Modelos Moleculares , Proteína Fosfatasa 2 , ARN Polimerasa II , ARN Polimerasa II/metabolismo , ARN Polimerasa II/química , ARN Polimerasa II/ultraestructura , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/ultraestructura , Terminación de la Transcripción Genética , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/química , Unión Proteica , Factores de Elongación Transcripcional/metabolismo , Factores de Elongación Transcripcional/química , Proteínas Nucleares/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/ultraestructura , Subunidades de Proteína/metabolismo , Subunidades de Proteína/química
13.
Nature ; 628(8006): 212-220, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509361

RESUMEN

RAD51 is the central eukaryotic recombinase required for meiotic recombination and mitotic repair of double-strand DNA breaks (DSBs)1,2. However, the mechanism by which RAD51 functions at DSB sites in chromatin has remained elusive. Here we report the cryo-electron microscopy structures of human RAD51-nucleosome complexes, in which RAD51 forms ring and filament conformations. In the ring forms, the N-terminal lobe domains (NLDs) of RAD51 protomers are aligned on the outside of the RAD51 ring, and directly bind to the nucleosomal DNA. The nucleosomal linker DNA that contains the DSB site is recognized by the L1 and L2 loops-active centres that face the central hole of the RAD51 ring. In the filament form, the nucleosomal DNA is peeled by the RAD51 filament extension, and the NLDs of RAD51 protomers proximal to the nucleosome bind to the remaining nucleosomal DNA and histones. Mutations that affect nucleosome-binding residues of the RAD51 NLD decrease nucleosome binding, but barely affect DNA binding in vitro. Consistently, yeast Rad51 mutants with the corresponding mutations are substantially defective in DNA repair in vivo. These results reveal an unexpected function of the RAD51 NLD, and explain the mechanism by which RAD51 associates with nucleosomes, recognizes DSBs and forms the active filament in chromatin.


Asunto(s)
Microscopía por Crioelectrón , Roturas del ADN de Doble Cadena , Nucleosomas , Recombinasa Rad51 , Proteínas de Saccharomyces cerevisiae , Humanos , ADN/química , ADN/metabolismo , ADN/ultraestructura , Reparación del ADN/genética , Nucleosomas/química , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Recombinasa Rad51/química , Recombinasa Rad51/metabolismo , Recombinasa Rad51/ultraestructura , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Mutación , Dominios Proteicos , Histonas/química , Histonas/metabolismo , Histonas/ultraestructura , Unión Proteica
14.
Nature ; 628(8008): 657-663, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509367

RESUMEN

In response to pathogen infection, gasdermin (GSDM) proteins form membrane pores that induce a host cell death process called pyroptosis1-3. Studies of human and mouse GSDM pores have revealed the functions and architectures of assemblies comprising 24 to 33 protomers4-9, but the mechanism and evolutionary origin of membrane targeting and GSDM pore formation remain unknown. Here we determine a structure of a bacterial GSDM (bGSDM) pore and define a conserved mechanism of pore assembly. Engineering a panel of bGSDMs for site-specific proteolytic activation, we demonstrate that diverse bGSDMs form distinct pore sizes that range from smaller mammalian-like assemblies to exceptionally large pores containing more than 50 protomers. We determine a cryo-electron microscopy structure of a Vitiosangium bGSDM in an active 'slinky'-like oligomeric conformation and analyse bGSDM pores in a native lipid environment to create an atomic-level model of a full 52-mer bGSDM pore. Combining our structural analysis with molecular dynamics simulations and cellular assays, our results support a stepwise model of GSDM pore assembly and suggest that a covalently bound palmitoyl can leave a hydrophobic sheath and insert into the membrane before formation of the membrane-spanning ß-strand regions. These results reveal the diversity of GSDM pores found in nature and explain the function of an ancient post-translational modification in enabling programmed host cell death.


Asunto(s)
Gasderminas , Myxococcales , Microscopía por Crioelectrón , Gasderminas/química , Gasderminas/metabolismo , Gasderminas/ultraestructura , Interacciones Hidrofóbicas e Hidrofílicas , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Simulación de Dinámica Molecular , Myxococcales/química , Myxococcales/citología , Myxococcales/ultraestructura , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteolisis , Piroptosis
15.
Photosynth Res ; 160(2-3): 61-75, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38488942

RESUMEN

The low-molecular-weight PsbM and PsbT proteins of Photosystem II (PS II) are both located at the monomer-monomer interface of the mature PS II dimer. Since the extrinsic proteins are associated with the final step of assembly of an active PS II monomer and, in the case of PsbO, are known to impact the stability of the PS II dimer, we have investigated the potential cooperativity between the PsbM and PsbT subunits and the PsbO, PsbU and PsbV extrinsic proteins. Blue-native polyacrylamide electrophoresis and western blotting detected stable PS II monomers in the ∆PsbM:∆PsbO and ∆PsbT:∆PsbO mutants that retained sufficient oxygen-evolving activity to support reduced photoautotrophic growth. In contrast, the ∆PsbM:∆PsbU and ∆PsbT:∆PsbU mutants assembled dimeric PS II at levels comparable to wild type and supported photoautotrophic growth at rates similar to those obtained with the corresponding ∆PsbM and ∆PsbT cells. Removal of PsbV was more detrimental than removal of PsbO. Only limited levels of dimeric PS II were observed in the ∆PsbM:∆PsbV mutant and the overall reduced level of assembled PS II in this mutant resulted in diminished rates of photoautotrophic growth and PS II activity below those obtained in the ∆PsbM:∆PsbO and ∆PsbT:∆PsbO strains. In addition, the ∆PsbT:∆PsbV mutant did not assemble active PS II centers although inactive monomers could be detected. The inability of the ∆PsbT:∆PsbV mutant to grow photoautotrophically, or to evolve oxygen, suggested a stable oxygen-evolving complex could not assemble in this mutant.


Asunto(s)
Complejo de Proteína del Fotosistema II , Synechocystis , Complejo de Proteína del Fotosistema II/metabolismo , Complejo de Proteína del Fotosistema II/genética , Synechocystis/metabolismo , Synechocystis/genética , Synechocystis/crecimiento & desarrollo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Mutación , Subunidades de Proteína/metabolismo , Oxígeno/metabolismo
16.
J Mol Graph Model ; 129: 108761, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38552302

RESUMEN

ADP-glucose pyrophosphorylase plays a pivotal role as an allosteric enzyme, essential for starch biosynthesis in plants. The higher plant AGPase comparises of a pair of large and a pair of small subunits to form a heterotetrameric complex. Growing evidence indicates that each subunit plays a distinct role in regulating the underlying mechanism of starch biosynthesis. In the rice genome, there are four large subunit genes (OsL1-L4) and three small subunit genes (OsS1, OsS2a, and OsS2b). While the structural assembly of cytosolic rice AGPase subunits (OsL2:OsS2b) has been elucidated, there is currently no such documented research available for plastidial rice AGPases (OsL1:OsS1). In this study, we employed protein modeling and MD simulation approaches to gain insights into the structural association of plastidial rice AGPase subunits. Our results demonstrate that the heterotetrameric association of OsL1:OsS1 is very similar to that of cytosolic OsL2:OsS2b and potato AGPase heterotetramer (StLS:StSS). Moreover, the yeast-two-hybrid results on OsL1:OsS1, which resemble StLS:StSS, suggest a differential protein assembly for OsL2:OsS2b. Thus, the regulatory and catalytic mechanisms for plastidial AGPases (OsL1:OsS1) could be different in rice culm and developing endosperm compared to those of OsL2:OsS2b, which are predominantly found in rice endosperm.


Asunto(s)
Oryza , Glucosa-1-Fosfato Adenililtransferasa/genética , Glucosa-1-Fosfato Adenililtransferasa/química , Glucosa-1-Fosfato Adenililtransferasa/metabolismo , Oryza/genética , Endospermo/genética , Endospermo/metabolismo , Simulación por Computador , Almidón/metabolismo , Subunidades de Proteína/metabolismo
17.
J Biol Chem ; 300(4): 107130, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432630

RESUMEN

The actin cytoskeleton and reactive oxygen species (ROS) both play crucial roles in various cellular processes. Previous research indicated a direct interaction between two key components of these systems: the WAVE1 subunit of the WAVE regulatory complex (WRC), which promotes actin polymerization and the p47phox subunit of the NADPH oxidase 2 complex (NOX2), which produces ROS. Here, using carefully characterized recombinant proteins, we find that activated p47phox uses its dual Src homology 3 domains to bind to multiple regions within the WAVE1 and Abi2 subunits of the WRC, without altering WRC's activity in promoting Arp2/3-mediated actin polymerization. Notably, contrary to previous findings, p47phox uses the same binding pocket to interact with both the WRC and the p22phox subunit of NOX2, albeit in a mutually exclusive manner. This observation suggests that when activated, p47phox may separately participate in two distinct processes: assembling into NOX2 to promote ROS production and engaging with WRC to regulate the actin cytoskeleton.


Asunto(s)
NADPH Oxidasa 2 , Familia de Proteínas del Síndrome de Wiskott-Aldrich , Humanos , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , NADPH Oxidasa 2/metabolismo , NADPH Oxidasa 2/genética , NADPH Oxidasas/metabolismo , NADPH Oxidasas/genética , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Sitios de Unión
18.
FEBS Lett ; 598(8): 875-888, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38553946

RESUMEN

Mammalian Ca2+-dependent Slo K+ channels can stably associate with auxiliary γ subunits which fundamentally alter their behavior. By a so far unknown mechanism, the four γ subunits reduce the need for voltage-dependent activation and, thereby, allow Slo to open independently of an action potential. Here, using cryo-EM, we reveal how the transmembrane helix of γ1/LRRC26 binds and presumably stabilizes the activated voltage-sensor domain of Slo1. The activation is further enhanced by an intracellular polybasic stretch which locally changes the charge gradient across the membrane. Our data provide a possible explanation for Slo1 regulation by the four γ subunits and also their different activation efficiencies. This suggests a novel activation mechanism of voltage-gated ion channels by auxiliary subunits.


Asunto(s)
Microscopía por Crioelectrón , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio , Subunidades de Proteína , Humanos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/química , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Subunidades de Proteína/metabolismo , Subunidades de Proteína/química , Animales , Activación del Canal Iónico , Modelos Moleculares , Células HEK293 , Unión Proteica , Dominios Proteicos
19.
Nat Commun ; 15(1): 2719, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38548794

RESUMEN

The study of phosphorylase kinase (PhK)-regulated glycogen metabolism has contributed to the fundamental understanding of protein phosphorylation; however, the molecular mechanism of PhK remains poorly understood. Here we present the high-resolution cryo-electron microscopy structures of human muscle PhK. The 1.3-megadalton PhK α4ß4γ4δ4 hexadecamer consists of a tetramer of tetramer, wherein four αßγδ modules are connected by the central ß4 scaffold. The α- and ß-subunits possess glucoamylase-like domains, but exhibit no detectable enzyme activities. The α-subunit serves as a bridge between the ß-subunit and the γδ subcomplex, and facilitates the γ-subunit to adopt an autoinhibited state. Ca2+-free calmodulin (δ-subunit) binds to the γ-subunit in a compact conformation. Upon binding of Ca2+, a conformational change occurs, allowing for the de-inhibition of the γ-subunit through a spring-loaded mechanism. We also reveal an ADP-binding pocket in the ß-subunit, which plays a role in allosterically enhancing PhK activity. These results provide molecular insights of this important kinase complex.


Asunto(s)
Músculos , Fosforilasa Quinasa , Humanos , Fosforilasa Quinasa/química , Fosforilasa Quinasa/metabolismo , Microscopía por Crioelectrón , Subunidades de Proteína/metabolismo , Músculos/metabolismo
20.
J Biol Chem ; 300(3): 105751, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38354779

RESUMEN

Eukaryotic DNA clamp is a trimeric protein featuring a toroidal ring structure that binds DNA on the inside of the ring and multiple proteins involved in DNA transactions on the outside. Eukaryotes have two types of DNA clamps: the replication clamp PCNA and the checkpoint clamp RAD9-RAD1-HUS1 (9-1-1). 9-1-1 activates the ATR-CHK1 pathway in DNA damage checkpoint, regulating cell cycle progression. Structure of 9-1-1 consists of two moieties: a hetero-trimeric ring formed by PCNA-like domains of three subunits and an intrinsically disordered C-terminal region of the RAD9 subunit, called RAD9 C-tail. The RAD9 C-tail interacts with the 9-1-1 ring and disrupts the interaction between 9-1-1 and DNA, suggesting a negative regulatory role for this intramolecular interaction. In contrast, RHINO, a 9-1-1 binding protein, interacts with both RAD1 and RAD9 subunits, positively regulating checkpoint activation by 9-1-1. This study presents a biochemical and structural analysis of intra- and inter-molecular interactions on the 9-1-1 ring. Biochemical analysis indicates that RAD9 C-tail binds to the hydrophobic pocket on the PCNA-like domain of RAD9, implying that the pocket is involved in multiple protein-protein interactions. The crystal structure of the 9-1-1 ring in complex with a RHINO peptide reveals that RHINO binds to the hydrophobic pocket of RAD9, shedding light on the RAD9-binding motif. Additionally, the study proposes a structural model of the 9-1-1-RHINO quaternary complex. Together, these findings provide functional insights into the intra- and inter-molecular interactions on the front side of RAD9, elucidating the roles of RAD9 C-tail and RHINO in checkpoint activation.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Complejos Multiproteicos , Subunidades de Proteína , Humanos , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , ADN/metabolismo , Daño del ADN , Reparación del ADN , Interacciones Hidrofóbicas e Hidrofílicas , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Dominios Proteicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA