Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Cell Sci ; 134(5)2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32501284

RESUMEN

Eukaryotic cell proliferation requires chromosome replication and precise segregation to ensure daughter cells have identical genomic copies. Species of the genus Plasmodium, the causative agents of malaria, display remarkable aspects of nuclear division throughout their life cycle to meet some peculiar and unique challenges to DNA replication and chromosome segregation. The parasite undergoes atypical endomitosis and endoreduplication with an intact nuclear membrane and intranuclear mitotic spindle. To understand these diverse modes of Plasmodium cell division, we have studied the behaviour and composition of the outer kinetochore NDC80 complex, a key part of the mitotic apparatus that attaches the centromere of chromosomes to microtubules of the mitotic spindle. Using NDC80-GFP live-cell imaging in Plasmodium berghei, we observe dynamic spatiotemporal changes during proliferation, including highly unusual kinetochore arrangements during sexual stages. We identify a very divergent candidate for the SPC24 subunit of the NDC80 complex, previously thought to be missing in Plasmodium, which completes a canonical, albeit unusual, NDC80 complex structure. Altogether, our studies reveal the kinetochore to be an ideal tool to investigate the non-canonical modes of chromosome segregation and cell division in Plasmodium.


Asunto(s)
Parásitos , Plasmodium , Animales , División Celular , Segregación Cromosómica/genética , Cinetocoros , Microtúbulos , Mitosis/genética , Plasmodium/genética , Huso Acromático/genética
2.
J Biol Chem ; 295(38): 13277-13286, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32723862

RESUMEN

The EAG (ether-à-go-go) family of voltage-gated K+ channels are important regulators of neuronal and cardiac action potential firing (excitability) and have major roles in human diseases such as epilepsy, schizophrenia, cancer, and sudden cardiac death. A defining feature of EAG (Kv10-12) channels is a highly conserved domain on the N terminus, known as the eag domain, consisting of a Per-ARNT-Sim (PAS) domain capped by a short sequence containing an amphipathic helix (Cap domain). The PAS and Cap domains are both vital for the normal function of EAG channels. Using heme-affinity pulldown assays and proteomics of lysates from primary cortical neurons, we identified that an EAG channel, hERG3 (Kv11.3), binds to heme. In whole-cell electrophysiology experiments, we identified that heme inhibits hERG3 channel activity. In addition, we expressed the Cap and PAS domain of hERG3 in Escherichia coli and, using spectroscopy and kinetics, identified the PAS domain as the location for heme binding. The results identify heme as a regulator of hERG3 channel activity. These observations are discussed in the context of the emerging role for heme as a regulator of ion channel activity in cells.


Asunto(s)
Corteza Cerebral/química , Canales de Potasio Éter-A-Go-Go/química , Hemo/química , Neuronas/química , Corteza Cerebral/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Hemo/metabolismo , Humanos , Neuronas/metabolismo , Unión Proteica , Dominios Proteicos
3.
Mol Microbiol ; 112(6): 1847-1862, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31562654

RESUMEN

Mycobacterium tuberculosis (Mtb) is able to persist in the body through months of multi-drug therapy. Mycobacteria possess a wide range of regulatory proteins, including the protein kinase B (PknB) which controls peptidoglycan biosynthesis during growth. Here, we observed that depletion of PknB resulted in specific transcriptional changes that are likely caused by reduced phosphorylation of the H-NS-like regulator Lsr2 at threonine 112. The activity of PknB towards this phosphosite was confirmed with purified proteins, and this site was required for adaptation of Mtb to hypoxic conditions, and growth on solid media. Like H-NS, Lsr2 binds DNA in sequence-dependent and non-specific modes. PknB phosphorylation of Lsr2 reduced DNA binding, measured by fluorescence anisotropy and electrophoretic mobility shift assays, and our NMR structure of phosphomimetic T112D Lsr2 suggests that this may be due to increased dynamics of the DNA-binding domain. Conversely, the phosphoablative T112A Lsr2 had increased binding to certain DNA sites in ChIP-sequencing, and Mtb containing this variant showed transcriptional changes that correspond with the change in DNA binding. In summary, PknB controls Mtb growth and adaptations to the changing host environment by phosphorylating the global transcriptional regulator Lsr2.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Mycobacterium tuberculosis/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Bacterianas/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Proteínas de Unión al ADN/fisiología , Ensayo de Cambio de Movilidad Electroforética/métodos , Regulación Bacteriana de la Expresión Génica/genética , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/fisiología , Treonina/metabolismo , Factores de Transcripción/metabolismo
4.
Anal Chem ; 91(15): 9516-9521, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31259536

RESUMEN

The growing significance of membrane proteins inspires continuous development and improvement of methods for robust membrane proteomics. Here, we developed a very simple and efficient method for membrane protein digestion using an ionic detergent, sodium dodecyl sulfate (SDS), at high temperature, conditions where trypsin is normally inactivated. Our results suggest that trypsin can be stabilized by a combination of calcium ions and sodium chloride, which enables protein digestion at elevated temperature in the presence of strong ionic detergents such as SDS. Finding the conditions for stabilization of trypsin offers novel opportunities for the application of detergents for the investigation of membrane proteins.


Asunto(s)
Calcio/química , Membrana Celular/química , Listeria monocytogenes/química , Ovalbúmina/química , Dodecil Sulfato de Sodio/química , Electroforesis en Gel de Poliacrilamida , Calor , Espectrometría de Masas , Reproducibilidad de los Resultados
5.
PLoS Pathog ; 13(5): e1006399, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28545104

RESUMEN

Sensing and response to changes in nutrient availability are essential for the lifestyle of environmental and pathogenic bacteria. Serine/threonine protein kinase G (PknG) is required for virulence of the human pathogen Mycobacterium tuberculosis, and its putative substrate GarA regulates the tricarboxylic acid cycle in M. tuberculosis and other Actinobacteria by protein-protein binding. We sought to understand the stimuli that lead to phosphorylation of GarA, and the roles of this regulatory system in pathogenic and non-pathogenic bacteria. We discovered that M. tuberculosis lacking garA was severely attenuated in mice and macrophages and furthermore that GarA lacking phosphorylation sites failed to restore the growth of garA deficient M. tuberculosis in macrophages. Additionally we examined the impact of genetic disruption of pknG or garA upon protein phosphorylation, nutrient utilization and the intracellular metabolome. We found that phosphorylation of GarA requires PknG and depends on nutrient availability, with glutamate and aspartate being the main stimuli. Disruption of pknG or garA caused opposing effects on metabolism: a defect in glutamate catabolism or depletion of intracellular glutamate, respectively. Strikingly, disruption of the phosphorylation sites of GarA was sufficient to recapitulate defects caused by pknG deletion. The results suggest that GarA is a cellular target of PknG and the metabolomics data demonstrate that the function of this signaling system is in metabolic regulation. This function in amino acid homeostasis is conserved amongst the Actinobacteria and provides an example of the close relationship between metabolism and virulence.


Asunto(s)
Aminoácidos/metabolismo , Proteínas Bacterianas/metabolismo , Metabolómica , Mycobacterium tuberculosis , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Ácido Aspártico/metabolismo , Proteínas Bacterianas/genética , Ácido Glutámico/metabolismo , Homeostasis , Macrófagos/microbiología , Ratones , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Tuberculosis/microbiología , Virulencia
6.
Int J Mol Sci ; 20(10)2019 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-31130675

RESUMEN

Site-specific incorporation of un-natural amino acids (UNAA) is a powerful approach to engineer and understand protein function. Site-specific incorporation of UNAAs is achieved through repurposing the amber codon (UAG) as a sense codon for the UNAA, using a tRNACUA that base pairs with an UAG codon in the mRNA and an orthogonal amino-acyl tRNA synthetase (aaRS) that charges the tRNACUA with the UNAA. Here, we report an expansion of the zebrafish genetic code to incorporate the UNAAs, azido-lysine (AzK), bicyclononyne-lysine (BCNK), and diazirine-lysine (AbK) into green fluorescent protein (GFP) and glutathione-s-transferase (GST). We also present proteomic evidence for UNAA incorporation into GFP. Our work sets the stage for the use of AzK, BCNK, and AbK introduction into proteins as a means to investigate and engineer their function in zebrafish.


Asunto(s)
Lisina/análogos & derivados , Ingeniería de Proteínas/métodos , Pez Cebra/genética , Animales , Codón de Terminación/genética , Código Genético , Glutatión Transferasa/genética , Proteínas Fluorescentes Verdes/genética , Lisina/genética , Proteínas de Pez Cebra/genética
8.
J Biol Chem ; 291(17): 8862-75, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26826123

RESUMEN

Establishing the in vivo activation status of G protein-coupled receptors would not only indicate physiological roles of G protein-coupled receptors but would also aid drug discovery by establishing drug/receptor engagement. Here, we develop a phospho-specific antibody-based biosensor to detect activation of the M1 muscarinic acetylcholine receptor (M1 mAChR) in vitro and in vivo Mass spectrometry phosphoproteomics identified 14 sites of phosphorylation on the M1 mAChR. Phospho-specific antibodies to four of these sites established that serine at position 228 (Ser(228)) on the M1 mAChR showed extremely low levels of basal phosphorylation that were significantly up-regulated by orthosteric agonist stimulation. In addition, the M1 mAChR-positive allosteric modulator, 1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid, enhanced acetylcholine-mediated phosphorylation at Ser(228) These data supported the hypothesis that phosphorylation at Ser(228) was an indicator of M1 mAChR activation. This was further supported in vivo by the identification of phosphorylated Ser(228) on the M1 mAChR in the hippocampus of mice following administration of the muscarinic ligands xanomeline and 1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid. Finally, Ser(228) phosphorylation was seen to increase in the CA1 region of the hippocampus following memory acquisition, a response that correlated closely with up-regulation of CA1 neuronal activity. Thus, determining the phosphorylation status of the M1 mAChR at Ser(228) not only provides a means of establishing receptor activation following drug treatment both in vitro and in vivo but also allows for the mapping of the activation status of the M1 mAChR in the hippocampus following memory acquisition thereby establishing a link between M1 mAChR activation and hippocampus-based memory and learning.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Técnicas Biosensibles/métodos , Región CA1 Hipocampal/metabolismo , Aprendizaje/fisiología , Memoria/fisiología , Fosfoproteínas/metabolismo , Receptor Muscarínico M1/metabolismo , Animales , Región CA1 Hipocampal/citología , Células CHO , Cricetinae , Cricetulus , Ratones , Fosfoproteínas/genética , Fosforilación/fisiología , Receptor Muscarínico M1/genética
9.
Biochem J ; 473(22): 4173-4192, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27623777

RESUMEN

The parathyroid hormone receptor 1 (PTH1R) is a member of family B of G-protein-coupled receptors (GPCRs), predominantly expressed in bone and kidney where it modulates extracellular Ca2+ homeostasis and bone turnover. It is well established that phosphorylation of GPCRs constitutes a key event in regulating receptor function by promoting arrestin recruitment and coupling to G-protein-independent signaling pathways. Mapping phosphorylation sites on PTH1R would provide insights into how phosphorylation at specific sites regulates cell signaling responses and also open the possibility of developing therapeutic agents that could target specific receptor functions. Here, we have used mass spectrometry to identify nine sites of phosphorylation in the C-terminal tail of PTH1R. Mutational analysis revealed identified two clusters of serine and threonine residues (Ser489-Ser495 and Ser501-Thr506) specifically responsible for the majority of PTH(1-34)-induced receptor phosphorylation. Mutation of these residues to alanine did not affect negatively on the ability of the receptor to couple to G-proteins or activate extracellular-signal-regulated kinase 1/2. Using fluorescence resonance energy transfer and bioluminescence resonance energy transfer to monitor PTH(1-34)-induced interaction of PTH1R with arrestin3, we show that the first cluster Ser489-Ser495 and the second cluster Ser501-Thr506 operated in concert to mediate both the efficacy and potency of ligand-induced arrestin3 recruitment. We further demonstrate that Ser503 and Thr504 in the second cluster are responsible for 70% of arrestin3 recruitment and are key determinants for interaction of arrestin with the receptor. Our data are consistent with the hypothesis that the pattern of C-terminal tail phosphorylation on PTH1R may determine the signaling outcome following receptor activation.


Asunto(s)
Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Secuencia de Aminoácidos , Arrestinas/metabolismo , Transferencia de Energía por Resonancia de Bioluminiscencia , Ensayo de Inmunoadsorción Enzimática , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Inmunoprecipitación , Espectrometría de Masas , Datos de Secuencia Molecular , Fosforilación , Receptor de Hormona Paratiroídea Tipo 1/química , Receptores Acoplados a Proteínas G/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal
10.
Proteomics ; 16(14): 1961-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27037516

RESUMEN

Unanchored polyubiquitin chains are emerging as important regulators of cellular physiology with diverse roles paralleling those of substrate-conjugated polyubiquitin. However tools able to discriminate unanchored polyubiquitin chains of different isopeptide linkages have not been reported. We describe the design of a linker-optimized ubiquitin-binding domain hybrid (t-UBD) containing two UBDs, a ZnF-UBP domain in tandem with a linkage-selective UBA domain, which exploits avidity effects to afford selective recognition of unanchored Lys48-linked polyubiquitin chains. Utilizing native MS to quantitatively probe binding affinities we confirm cooperative binding of the UBDs within the synthetic protein, and desired binding specificity for Lys48-linked ubiquitin dimers. Furthermore, MS/MS analyses indicate that the t-UBD, when applied as an affinity enrichment reagent, can be used to favor the purification of endogenous unanchored Lys48-linked polyubiquitin chains from mammalian cell extracts. Our study indicates that strategies for the rational design and engineering of polyubiquitin chain-selective binding in nonbiological polymers are possible, paving the way for the generation of reagents to probe unanchored polyubiquitin chains of different linkages and more broadly the 'ubiquitome'. All MS data have been deposited in the ProteomeXchange with identifier PXD004059 (http://proteomecentral.proteomexchange.org/dataset/PXD004059).


Asunto(s)
Bioensayo/normas , Lisina/metabolismo , Poliubiquitina/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Sitios de Unión , Mezclas Complejas/química , Expresión Génica , Células HEK293 , Humanos , Cinética , Lisina/química , Modelos Moleculares , Poliubiquitina/química , Unión Proteica , Dominios Proteicos , Ingeniería de Proteínas , Multimerización de Proteína , Proteínas Recombinantes de Fusión/genética , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem , Ubiquitinación
11.
Malar J ; 15(1): 535, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27821169

RESUMEN

BACKGROUND: Examining essential biochemical pathways in Plasmodium falciparum presents serious challenges, as standard molecular techniques such as siRNA cannot be employed in this organism, and generating gene knock-outs of essential proteins requires specialized conditional approaches. In the study of protein kinases, pharmacological inhibition presents a feasible alternative option. However, as in mammalian systems, inhibitors often lack the desired selectivity. Described here is a chemical genetic approach to selectively inhibit Pfnek-2 in P. falciparum, a member of the NIMA-related kinase family that is essential for completion of the sexual development of the parasite. RESULTS: Introduction of a valine to cysteine mutation at position 24 in the glycine rich loop of Pfnek-2 does not affect kinase activity but confers sensitivity to the protein kinase inhibitor 4-(6-ethynyl-9H-purin-2-ylamino) benzene sulfonamide (NCL-00016066). Using a combination of in vitro kinase assays and mass spectrometry, (including phosphoproteomics) the study shows that this compound acts as an irreversible inhibitor to the mutant Pfnek2 likely through a covalent link with the introduced cysteine residue. In particular, this was shown by analysis of total protein mass using mass spectrometry which showed a shift in molecular weight of the mutant kinase in the presence of the inhibitor to be precisely equivalent to the molecular weight of NCL-00016066. A similar molecular weight shift was not observed in the wild type kinase. Importantly, this inhibitor has little activity towards the wild type Pfnek-2 and, therefore, has all the properties of an effective chemical genetic tool that could be employed to determine the cellular targets for Pfnek-2. CONCLUSIONS: Allelic replacement of wild-type Pfnek-2 with the mutated kinase will allow for targeted inhibition of Pfnek-2 with NCL-00016066 and hence pave the way for comparative studies aimed at understanding the biological role and transmission-blocking potential of Pfnek-2.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Proteínas Mutantes/metabolismo , Quinasas Relacionadas con NIMA/metabolismo , Plasmodium falciparum/enzimología , Purinas/metabolismo , Sulfonamidas/metabolismo , Espectrometría de Masas , Proteínas Mutantes/genética , Quinasas Relacionadas con NIMA/genética
12.
bioRxiv ; 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36778504

RESUMEN

Mechanisms of cell division are remarkably diverse, suggesting the underlying molecular networks among eukaryotes differ extensively. The Aurora family of kinases orchestrates the process of chromosome segregation and cytokinesis during cell division through precise spatiotemporal regulation of their catalytic activities by distinct scaffolds. Plasmodium spp., the causative agents of malaria, are unicellular eukaryotes that have three divergent aurora-related kinases (ARKs) and lack most canonical scaffolds/activators. The parasite uses unconventional modes of chromosome segregation during endomitosis and meiosis in sexual transmission stages within mosquito host. This includes a rapid threefold genome replication from 1N to 8N with successive cycles of closed mitosis, spindle formation and chromosome segregation within eight minutes (termed male gametogony). Kinome studies had previously suggested likely essential functions for all three Plasmodium ARKs during asexual mitotic cycles; however, little is known about their location, function, or their scaffolding molecules during unconventional sexual proliferative stages. Using a combination of super-resolution microscopy, mass spectrometry, and live-cell fluorescence imaging, we set out to investigate the role of the atypical Aurora paralog ARK2 to proliferative sexual stages using rodent malaria model Plasmodium berghei . We find that ARK2 primarily localises to the spindle apparatus in the vicinity of kinetochores during both mitosis and meiosis. Interactomics and co-localisation studies reveal a unique ARK2 scaffold at the spindle including the microtubule plus end-binding protein EB1, lacking conserved Aurora scaffold proteins. Gene function studies indicate complementary functions of ARK2 and EB1 in driving endomitotic divisions and thereby parasite transmission. Our discovery of a novel Aurora kinase spindle scaffold underlines the emerging flexibility of molecular networks to rewire and drive unconventional mechanisms of chromosome segregation in the malaria parasite Plasmodium .

13.
Res Sq ; 2023 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-36798191

RESUMEN

Mechanisms of cell division are remarkably diverse, suggesting the underlying molecular networks among eukaryotes differ extensively. The Aurora family of kinases orchestrates the process of chromosome segregation and cytokinesis during cell division through precise spatiotemporal regulation of their catalytic activities by distinct scaffolds. Plasmodium spp., the causative agents of malaria, are unicellular eukaryotes that have three divergent aurora-related kinases (ARKs) and lack most canonical scaffolds/activators. The parasite uses unconventional modes of chromosome segregation during endomitosis and meiosis in sexual transmission stages within mosquito host. This includes a rapid threefold genome replication from 1N to 8N with successive cycles of closed mitosis, spindle formation and chromosome segregation within eight minutes (termed male gametogony). Kinome studies had previously suggested likely essential functions for all three Plasmodium ARKs during asexual mitotic cycles; however, little is known about their location, function, or their scaffolding molecules during unconventional sexual proliferative stages. Using a combination of super-resolution microscopy, mass spectrometry, omics and live-cell fluorescence imaging, we set out to investigate the contribution of the atypical Aurora paralog ARK2 to proliferative sexual stages using rodent malaria model Plasmodium berghei. We find that ARK2 primarily localises to the spindle apparatus associated with kinetochores during both mitosis and meiosis. Interactomics and co-localisation studies reveal a unique ARK2 scaffold at the spindle including the microtubule plus end-binding protein EB1 and lacking some other conserved molecules. Gene function studies indicate complementary functions of ARK2 and EB1 in driving endomitotic divisions and thereby parasite transmission. Our discovery of a novel Aurora spindle scaffold underlines the emerging flexibility of molecular networks to rewire and drive unconventional mechanisms of chromosome segregation in the malaria parasite Plasmodium.

14.
Nat Commun ; 14(1): 5652, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37704606

RESUMEN

The Aurora family of kinases orchestrates chromosome segregation and cytokinesis during cell division, with precise spatiotemporal regulation of its catalytic activities by distinct protein scaffolds. Plasmodium spp., the causative agents of malaria, are unicellular eukaryotes with three unique and highly divergent aurora-related kinases (ARK1-3) that are essential for asexual cellular proliferation but lack most canonical scaffolds/activators. Here we investigate the role of ARK2 during sexual proliferation of the rodent malaria Plasmodium berghei, using a combination of super-resolution microscopy, mass spectrometry, and live-cell fluorescence imaging. We find that ARK2 is primarily located at spindle microtubules in the vicinity of kinetochores during both mitosis and meiosis. Interactomic and co-localisation studies reveal several putative ARK2-associated interactors including the microtubule-interacting protein EB1, together with MISFIT and Myosin-K, but no conserved eukaryotic scaffold proteins. Gene function studies indicate that ARK2 and EB1 are complementary in driving endomitotic division and thereby parasite transmission through the mosquito. This discovery underlines the flexibility of molecular networks to rewire and drive unconventional mechanisms of chromosome segregation in the malaria parasite.


Asunto(s)
División del Núcleo Celular , Segregación Cromosómica , Animales , Plasmodium berghei/genética , Proliferación Celular , Meiosis , Aurora Quinasas , Eucariontes
15.
J Neurochem ; 123(5): 725-35, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22971236

RESUMEN

Recombinant FlagHis(6) tagged Human P2X1 receptors expressed in HEK293 cells were purified, digested with trypsin and analysed by mass spectroscopy (96% coverage following de-glycosylation and reduction). The receptor was basally phosphorylated at residues S387, S388 and T389 in the carboxyl terminus, a triple alanine mutant of these residues had a modest ~ 25% increase in current amplitude and recovery from desensitization. Chemical modification showed that intracellular lysine residues close to the transmembrane domains and the membrane stabilization motif are accessible to the aqueous environment. The membrane-impermeant cross-linking reagent 3,3'-Dithiobis (sulfosuccinimidylpropionate) (DTSSP) reduced agonist binding and P2X1 receptor currents by > 90%, and modified lysine residues were identified by mass spectroscopy. Mutation to remove reactive lysine residues around the ATP-binding pocket had no effect on inhibtion of agonist evoked currents following DTSSP. However, agonist evoked currents were ~ 10-fold higher than for wild type following DTSSP treatment for mutants K199R, K221R and K199R-K221R. These mutations remove reactive residues distant from the agonist binding pocket that are close enough to cross-link adjacent subunits. These results suggest that conformational change in the P2X1 receptor is required for co-ordination of ATP action.


Asunto(s)
Receptores Purinérgicos P2X1/química , Receptores Purinérgicos P2X1/metabolismo , Transducción de Señal/fisiología , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Potenciales Evocados/fisiología , Células HEK293 , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Estructura Cuaternaria de Proteína , Espectrometría de Masas en Tándem , Xenopus laevis
16.
Sci Signal ; 15(760): eabm3720, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36378750

RESUMEN

Many dementias are propagated through the spread of "prion-like" misfolded proteins. This includes prion diseases themselves (such as Creutzfeldt-Jakob disease) and Alzheimer's disease (AD), for which no treatments are available to slow or stop progression. The M1 acetylcholine muscarinic receptor (M1 receptor) is abundant in the brain, and its activity promotes cognitive function in preclinical models and in patients with AD. Here, we investigated whether activation of the M1 receptor might slow the progression of neurodegeneration associated with prion-like misfolded protein in a mouse model of prion disease. Proteomic and transcriptomic analysis of the hippocampus revealed that this model had a molecular profile that was similar to that of human neurodegenerative diseases, including AD. Chronic enhancement of the activity of the M1 receptor with the positive allosteric modulator (PAM) VU0486846 reduced the abundance of prion-induced molecular markers of neuroinflammation and mitochondrial dysregulation in the hippocampus and normalized the abundance of those associated with neurotransmission, including synaptic and postsynaptic signaling components. PAM treatment of prion-infected mice prolonged survival and maintained cognitive function. Thus, allosteric activation of M1 receptors may reduce the severity of neurodegenerative diseases caused by the prion-like propagation of misfolded protein.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Enfermedades por Prión , Priones , Humanos , Animales , Ratones , Priones/genética , Enfermedades Neurodegenerativas/genética , Patología Molecular , Proteómica , Enfermedades por Prión/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo
17.
Cell Surf ; 7: 100052, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34296047

RESUMEN

Mycobacterium tuberculosis (Mtb) is an intracellular human pathogen that has evolved to survive in a nutrient limited environment within the host for decades. Accordingly, Mtb has developed strategies to acquire scarce nutrients and the mycobacterial transporter systems provide an important route for the import of key energy sources. However, the physiological role of the Mtb transporters and their substrate preference(s) are poorly characterised. Previous studies have established that the Mtb UspC solute-binding domain recognises amino- and phosphorylated-sugars, indicating that the mycobacterial UspABC transporter plays a key role in the import of peptidoglycan precursors. Herein, we have used a wide array of approaches to investigate the role of UspABC in Mycobacterium smegmatis by analysis of mutant strains that either lack the solute binding domain: ΔuspC or the entire transport complex: ΔuspABC. Analysis of mycobacterial transcripts shows that the uspABC system is functionally expressed in mycobacteria as a contiguous reading frame. Topology mapping confirms an Nin-Cin orientation of the UspAB integral membrane spanning domains. Phenotypic microarray profiling of commercially available sugars suggests, unexpectedly, that the uspC and ΔuspABC mutants had different carbon utilisation profiles and that neither strain utilised glucose-1-phosphate. Furthermore, proteomics analysis showed an alteration in the abundance of proteins involved in sugar and lipid metabolism, crucial for cell envelope synthesis, and we propose that UspABC has an important role in determining the interplay between these pathways.

18.
Nat Commun ; 12(1): 530, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33483501

RESUMEN

The emergence and spread of artemisinin resistance, driven by mutations in Plasmodium falciparum K13, has compromised antimalarial efficacy and threatens the global malaria elimination campaign. By applying systems-based quantitative transcriptomics, proteomics, and metabolomics to a panel of isogenic K13 mutant or wild-type P. falciparum lines, we provide evidence that K13 mutations alter multiple aspects of the parasite's intra-erythrocytic developmental program. These changes impact cell-cycle periodicity, the unfolded protein response, protein degradation, vesicular trafficking, and mitochondrial metabolism. K13-mediated artemisinin resistance in the Cambodian Cam3.II line was reversed by atovaquone, a mitochondrial electron transport chain inhibitor. These results suggest that mitochondrial processes including damage sensing and anti-oxidant properties might augment the ability of mutant K13 to protect P. falciparum against artemisinin action by helping these parasites undergo temporary quiescence and accelerated growth recovery post drug elimination.


Asunto(s)
Artemisininas/farmacología , Resistencia a Medicamentos/genética , Eritrocitos/metabolismo , Mutación , Plasmodium falciparum/genética , Antimaláricos/farmacología , Atovacuona/farmacología , Puntos de Control del Ciclo Celular/genética , Eritrocitos/parasitología , Perfilación de la Expresión Génica/métodos , Humanos , Metabolómica/métodos , Mitocondrias/genética , Mitocondrias/metabolismo , Modelos Genéticos , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiología , Proteómica/métodos , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
19.
Commun Biol ; 4(1): 760, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34145386

RESUMEN

PP1 is a conserved eukaryotic serine/threonine phosphatase that regulates many aspects of mitosis and meiosis, often working in concert with other phosphatases, such as CDC14 and CDC25. The proliferative stages of the malaria parasite life cycle include sexual development within the mosquito vector, with male gamete formation characterized by an atypical rapid mitosis, consisting of three rounds of DNA synthesis, successive spindle formation with clustered kinetochores, and a meiotic stage during zygote to ookinete development following fertilization. It is unclear how PP1 is involved in these unusual processes. Using real-time live-cell and ultrastructural imaging, conditional gene knockdown, RNA-seq and proteomic approaches, we show that Plasmodium PP1 is implicated in both mitotic exit and, potentially, establishing cell polarity during zygote development in the mosquito midgut, suggesting that small molecule inhibitors of PP1 should be explored for blocking parasite transmission.


Asunto(s)
Estadios del Ciclo de Vida/genética , Meiosis/genética , Mitosis/genética , Plasmodium/crecimiento & desarrollo , Proteína Fosfatasa 1/genética , Proteínas Protozoarias/genética , Proliferación Celular/genética , Malaria/prevención & control , Malaria/transmisión , Mosquitos Vectores/parasitología , Plasmodium/metabolismo , Proteína Fosfatasa 1/metabolismo , Proteínas Protozoarias/metabolismo
20.
J Biol Chem ; 284(25): 17147-17156, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19332541

RESUMEN

Changes in synaptic strength mediated by ionotropic glutamate N-methyl-D-asparate (NMDA) receptors is generally considered to be the molecular mechanism underlying memory and learning. NMDA receptors themselves are subject to regulation through signaling pathways that are activated by G-protein-coupled receptors (GPCRs). In this study we investigate the ability of NMDA receptors to regulate the signaling of GPCRs by focusing on the G(q/11)-coupled M(3)-muscarinic receptor expressed endogenously in mouse cerebellar granule neurons. We show that NMDA receptor activation results in the phosphorylation and desensitization of M(3)-muscarinic receptors through a mechanism dependent on NMDA-mediated calcium influx and the activity of calcium-calmodulin-dependent protein kinase II. Our study reveals a complex pattern of regulation where GPCRs (M(3)-muscarinic) and NMDA receptors can feedback on each other in a process that is likely to influence the threshold value of signaling networks involved in synaptic plasticity.


Asunto(s)
Cerebelo/metabolismo , Receptor Muscarínico M3/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Secuencia de Aminoácidos , Animales , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Cerebelo/citología , Retroalimentación Fisiológica , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , N-Metilaspartato/farmacología , Plasticidad Neuronal , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosfatidilinositoles/metabolismo , Fosforilación , Receptor Muscarínico M3/química , Receptor Muscarínico M3/deficiencia , Receptor Muscarínico M3/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda