Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 59
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Proc Natl Acad Sci U S A ; 121(26): e2401154121, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38889150

RÉSUMÉ

Almost all elongator tRNAs (Transfer RNAs) harbor 5-methyluridine 54 and pseudouridine 55 in the T arm, generated by the enzymes TrmA and TruB, respectively, in Escherichia coli. TrmA and TruB both act as tRNA chaperones, and strains lacking trmA or truB are outcompeted by wild type. Here, we investigate how TrmA and TruB contribute to cellular fitness. Deletion of trmA and truB in E. coli causes a global decrease in aminoacylation and alters other tRNA modifications such as acp3U47. While overall protein synthesis is not affected in ΔtrmA and ΔtruB strains, the translation of a subset of codons is significantly impaired. As a consequence, we observe translationally reduced expression of many specific proteins, that are either encoded with a high frequency of these codons or that are large proteins. The resulting proteome changes are not related to a specific growth phenotype, but overall cellular fitness is impaired upon deleting trmA and truB in accordance with a general protein synthesis impact. In conclusion, we demonstrate that universal modifications of the tRNA T arm are critical for global tRNA function by enhancing tRNA maturation, tRNA aminoacylation, and translation, thereby improving cellular fitness irrespective of the growth conditions which explains the conservation of trmA and truB.


Sujet(s)
Escherichia coli , ARN de transfert , ARN de transfert/métabolisme , ARN de transfert/génétique , Escherichia coli/génétique , Escherichia coli/métabolisme , Biosynthèse des protéines , Protéines Escherichia coli/métabolisme , Protéines Escherichia coli/génétique , T-RNA methyltransferases/métabolisme , T-RNA methyltransferases/génétique , Maturation post-transcriptionnelle des ARN
2.
Sci Rep ; 13(1): 21229, 2023 12 01.
Article de Anglais | MEDLINE | ID: mdl-38040826

RÉSUMÉ

Myasthenia Gravis (MG) is an autoimmune disease associated with severe neuromuscular weakness. Diagnostic confirmation of MG is typically delayed and secured in about 85% and 50% of patients with generalized and ocular MG, respectively with serum antibodies. We have identified a sensitive and specific diagnostic biomarker for various MG serotypes with quantitative proteomics. Serum proteomes of 18 individuals (MG patients, healthy controls (HC), Rheumatoid Arthritis (RA) were quantified in a pilot study and occurrence of high residual fibrinogen was validated by immunoblotting and further investigated by targeted mass spectrometry on the sera of 79 individuals (31 MG of various serotypes, 30 HC, 18 RA). Initial proteomic analysis identified high residual fibrinogen in MG patient sera which was then validated by antibody-based testing. Subsequently, a blinded study of independent samples showed 100% differentiation of MG patients from controls. A final serological quantification of 14 surrogate peptides derived from α-, ß-, and γ-subunits of fibrinogen in 79 individuals revealed fibrinogen to be highly specific and 100% sensitive for MG (p < 0.00001), with a remarkable average higher abundance of > 1000-fold over control groups. Our unanticipated discovery of high levels of residual serum fibrinogen in all MG patients can secure rapid bedside diagnosis of MG.


Sujet(s)
Polyarthrite rhumatoïde , Hémostatiques , Myasthénie , Humains , Fibrinogène , Protéomique , Projets pilotes , Sérogroupe , Marqueurs biologiques , Autoanticorps
3.
Cell Signal ; 110: 110830, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37516395

RÉSUMÉ

Cellular signalling cues lead to the initiation of apoptotic pathways and often result in the activation of caspases which in turn cause the generation of proteolytically generated protein fragments with new or altered functions. Mounting number of studies reveal that the activity of these proteolytically activated protein fragments can be counteracted via their selective degradation by the N-degron degradation pathways. Here, we investigate the proteolytically generated fragment of the PKC theta kinase, where we demonstrate the first report on the stability of this pro-apoptotic protein fragment. We have determined that the pro-apoptotic cleaved fragment of PKC-theta is unstable in cells because its N-terminal lysine targets it for proteasomal degradation via the N-degron degradation pathway and this degradation is inhibited by mutating the destabilizing N-termini, knockdown of the UBR1 and UBR2 E3 ligases. Tellingly, we demonstrate that the metabolic stabilization of the cleaved fragment of PKC-theta or inhibition of the N-degron degradation augments the apoptosis-inducing effect of staurosporine in Jurkat cells. Notably, we have unveiled that the cleaved fragment of PKC theta, per se, can induce apoptotic cell death in Jurkat T-cell leukemia. Our results expand the functional scope of mammalian N-degron degradation pathways, and support the notion that targeting N-degron degradation machinery may have promising therapeutic implications in cancer cells.


Sujet(s)
Caspases , Ubiquitin-protein ligases , Animaux , Humains , Protein Kinase C-theta/métabolisme , Caspases/métabolisme , Ubiquitin-protein ligases/métabolisme , Apoptose , Cellules Jurkat , Protéolyse , Mammifères/métabolisme
4.
J Magn Reson ; 353: 107499, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37307676

RÉSUMÉ

Solution NMR studies of large proteins are hampered by rapid signal decay due to short-range dipolar 1H-1H and 1H-13C interactions. These are attenuated by rapid rotation in methyl groups and by deuteration (2H), so selective 1H,13C-isotope labelling of methyl groups in otherwise perdeuterated proteins, combined with methyl transverse relaxation optimized spectroscopy (methyl-TROSY), is now standard for solution NMR of large protein systems > 25 kDa. For non-methyl positions, long-lived magnetization can be introduced as isolated 1H-12C groups. We have developed a cost-effective chemical synthesis for producing selectively deuterated phenylpyruvate and hydroxyphenylpyruvate. Feeding these amino acid precursors to E. coli in D2O, along with selectively deuterated anthranilate and unlabeled histidine, results in isolated and long-lived 1H magnetization in the aromatic rings of Phe (HD, HZ), Tyr (HD), Trp (HH2, HE3) and His (HD2 and HE1). We are additionally able to obtain stereoselective deuteration of Asp, Asn, and Lys amino acid residues using unlabeled glucose and fumarate as carbon sources and oxalate and malonate as metabolic inhibitors. Combining these approaches produces isolated 1H-12C groups in Phe, Tyr, Trp, His, Asp, Asn, and Lys in a perdeuterated background, which is compatible with standard 1H-13C labeling of methyl groups in Ala, Ile, Leu, Val, Thr, Met. We show that isotope labeling of Ala is improved using the transaminase inhibitor L-cycloserine, and labeling of Thr is improved through addition of Cys and Met, which are known inhibitors of homoserine dehydrogenase. We demonstrate the creation of long-lived 1H NMR signals in most amino acid residues using our model system, the WW domain of human Pin1, as well as the bacterial outer membrane protein PagP.


Sujet(s)
Protéines Escherichia coli , Escherichia coli , Humains , Analyse coût-bénéfice , Spectroscopie par résonance magnétique du proton , Acides aminés aromatiques , Acides aminés , Acyltransferases
5.
J Biol Chem ; 298(5): 101925, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35413287

RÉSUMÉ

Conditional proteolytic degradation is an irreversible and highly regulated process that fulfills crucial regulatory functions in all organisms. As proteolytic targets tend to be critical metabolic or regulatory proteins, substrates are targeted for degradation only under appropriate conditions through the recognition of an amino acid sequence referred to as a "degron". DEAD-box RNA helicases mediate all aspects of RNA metabolism, contributing to cellular fitness. However, the mechanism by which abiotic-stress modulation of protein stability regulates bacterial helicase abundance has not been extensively characterized. Here, we provide in vivo evidence that proteolytic degradation of the cyanobacterial DEAD-box RNA helicase CrhR is conditional, being initiated by a temperature upshift from 20 to 30 °C in the model cyanobacterium, Synechocystis sp. PCC 6803. We show degradation requires a unique, highly conserved, inherently bipartite degron located in the C-terminal extension found only in CrhR-related RNA helicases in the phylum Cyanobacteria. However, although necessary, the degron is not sufficient for proteolysis, as disruption of RNA helicase activity and/or translation inhibits degradation. These results suggest a positive feedback mechanism involving a role for CrhR in expression of a crucial factor required for degradation. Furthermore, AlphaFold structural prediction indicated the C-terminal extension is a homodimerization domain with homology to other bacterial RNA helicases, and mass photometry data confirmed that CrhR exists as a dimer in solution at 22 °C. These structural data suggest a model wherein the CrhR degron is occluded at the dimerization interface but could be exposed if dimerization was disrupted by nonpermissive conditions.


Sujet(s)
DEAD-box RNA helicases , Synechocystis , DEAD-box RNA helicases/métabolisme , Protéolyse , ARN bactérien/métabolisme , Synechocystis/enzymologie , Synechocystis/génétique
6.
Neurotox Res ; 40(1): 298-318, 2022 Feb.
Article de Anglais | MEDLINE | ID: mdl-35043375

RÉSUMÉ

Among the most salient features that underpin the development of aging-related neurodegenerative disorders are the accumulation of protein aggregates and the decrease in cellular degradation capacity. Mammalian cells have evolved sophisticated quality control mechanisms to repair or eliminate the otherwise abnormal or misfolded proteins. Chaperones identify unstable or abnormal conformations in proteins and often help them regain their correct conformation. However, if repair is not an option, abnormal proteins are selectively degraded to prevent undesired interactions with other proteins or oligomerization into toxic multimeric complexes. The autophagic-lysosomal system and the ubiquitin-proteasome system mediate the selective and targeted degradation of abnormal or aberrant protein fragments. Despite an increasing understanding regarding the molecular responses that counteract the formation and clearance of dysfunctional protein aggregates, the role of N-degrons in these processes is poorly understood. Previous work demonstrated that the Arg-N-end rule degradation pathway (Arg-N-degron pathway) mediates the degradation of neurodegeneration-associated proteins, thereby regulating crucial signaling hubs that modulate the progression of neurodegenerative diseases. Herein, we discuss the functional interconnection between N-degron pathways and proteins associated with neurodegenerative disorders, including Alzheimer's disease, amyotrophic lateral sclerosis, and Parkinson's disease. We also highlight some future prospects related to how the molecular insights gained from these processes will help unveil novel therapeutic approaches.


Sujet(s)
Maladies neurodégénératives , Ubiquitine , Animaux , Mammifères/métabolisme , Chaperons moléculaires/métabolisme , Maladies neurodégénératives/métabolisme , Proteasome endopeptidase complex/métabolisme , Protéolyse , Ubiquitine/métabolisme
7.
J Biol Chem ; 297(2): 100959, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34265304

RÉSUMÉ

MAGEL2 encodes the L2 member of the melanoma-associated antigen gene (MAGE) protein family, truncating mutations of which can cause Schaaf-Yang syndrome, an autism spectrum disorder. MAGEL2 is also inactivated in Prader-Willi syndrome, which overlaps clinically and mechanistically with Schaaf-Yang syndrome. Studies to date have only investigated the C-terminal portion of the MAGEL2 protein, containing the MAGE homology domain that interacts with RING-E3 ubiquitin ligases and deubiquitinases to form protein complexes that modify protein ubiquitination. In contrast, the N-terminal portion of the MAGEL2 protein has never been studied. Here, we find that MAGEL2 has a low-complexity intrinsically disordered N-terminus rich in Pro-Xn-Gly motifs that is predicted to mediate liquid-liquid phase separation to form biomolecular condensates. We used proximity-dependent biotin identification (BioID) and liquid chromatography-tandem mass spectrometry to identify MAGEL2-proximal proteins, then clustered these proteins into functional networks. We determined that coding mutations analogous to disruptive mutations in other MAGE proteins alter these networks in biologically relevant ways. Proteins identified as proximal to the N-terminal portion of MAGEL2 are primarily involved in mRNA metabolic processes and include three mRNA N 6-methyladenosine (m6A)-binding YTHDF proteins and two RNA interference-mediating TNRC6 proteins. We found that YTHDF2 coimmunoprecipitates with MAGEL2, and coexpression of MAGEL2 reduces the nuclear accumulation of YTHDF2 after heat shock. We suggest that the N-terminal region of MAGEL2 may have a role in RNA metabolism and in particular the regulation of mRNAs modified by m6A methylation. These results provide mechanistic insight into pathogenic MAGEL2 mutations associated with Schaaf-Yang syndrome and related disorders.


Sujet(s)
Syndrome de Prader-Willi , Protéines/composition chimique , Protéines/métabolisme , ARN/métabolisme , Humains , Mutation , Phénotype , Domaines protéiques
8.
Front Genet ; 12: 689922, 2021.
Article de Anglais | MEDLINE | ID: mdl-34234816

RÉSUMÉ

Molecular chaperone networks fulfill complex roles in protein homeostasis and are essential for maintaining cell health. Hsp40s (commonly referred to as J-proteins) have critical roles in development and are associated with a variety of human diseases, yet little is known regarding the J-proteins with respect to the post-transcriptional mechanisms that regulate their expression. With relatively small alterations in their abundance and stoichiometry altering their activity, post-transcriptional regulation potentially has significant impact on the functions of J-proteins. MicroRNAs (miRNAs) are a large group of non-coding RNAs that form a complex regulatory network impacting gene expression. Here we review and investigate the current knowledge and potential intersection of miRNA regulatory networks with the J-Protein chaperone network. Analysis of datasets from the current version of TargetScan revealed a great number of predicted microRNAs targeting J-proteins compared to the limited reports of interactions to date. There are likely unstudied regulatory interactions that influence chaperone biology contained within our analysis. We go on to present some criteria for prioritizing candidate interactions including potential cooperative targeting of J-Proteins by multiple miRNAs. In summary, we offer a view on the scope of regulation of J-Proteins through miRNAs with the aim of guiding future investigations by identifying key regulatory nodes within these two complex cellular networks.

9.
Nat Commun ; 12(1): 2939, 2021 05 19.
Article de Anglais | MEDLINE | ID: mdl-34011960

RÉSUMÉ

Elucidation of non-canonical protein functions can identify novel tissue homeostasis pathways. Herein, we describe a role for the Bcl-2 family member BAD in postnatal mammary gland morphogenesis. In Bad3SA knock-in mice, where BAD cannot undergo phosphorylation at 3 key serine residues, pubertal gland development is delayed due to aberrant tubulogenesis of the ductal epithelium. Proteomic and RPPA analyses identify that BAD regulates focal adhesions and the mRNA translation repressor, 4E-BP1. These results suggest that BAD modulates localized translation that drives focal adhesion maturation and cell motility. Consistent with this, cells within Bad3SA organoids contain unstable protrusions with decreased compartmentalized mRNA translation and focal adhesions, and exhibit reduced cell migration and tubulogenesis. Critically, protrusion stability is rescued by 4E-BP1 depletion. Together our results confirm an unexpected role of BAD in controlling localized translation and cell migration during mammary gland development.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Protéines du cycle cellulaire/métabolisme , Glandes mammaires animales/croissance et développement , Glandes mammaires animales/métabolisme , Glandes mammaires humaines/croissance et développement , Glandes mammaires humaines/métabolisme , Protéine Bad/métabolisme , Substitution d'acide aminé , Animaux , Lignée cellulaire , Mouvement cellulaire/génétique , Femelle , Techniques de knock-in de gènes , Humains , Souris , Souris de lignée C57BL , Souris knockout , Modèles animaux , Morphogenèse , Protéines mutantes/composition chimique , Protéines mutantes/génétique , Protéines mutantes/métabolisme , Organoïdes/croissance et développement , Organoïdes/métabolisme , Phosphorylation , Biosynthèse des protéines , ARN messager/génétique , ARN messager/métabolisme , Sérine/composition chimique , Protéine Bad/déficit , Protéine Bad/génétique
10.
Endocrinology ; 161(12)2020 12 01.
Article de Anglais | MEDLINE | ID: mdl-33159513

RÉSUMÉ

In mammals, protein degradation is mediated selectively by the ubiquitin proteasome system (UPS) and the autophagic-lysosomal system. Over the past decades, N-degron pathways have been shown to be responsible for the selective degradation of proteins that harbor destabilizing N-terminal motifs. Recent studies have employed these pathways in the development of proteolysis targeting chimeras (PROTACs) composed of a degradation module linked to a substrate recognition domain to target proteins encoded by cancer-related genes for proteasomal destruction. Herein we provide an overview of PROTACs in the context of the N-degron concept and address the application of this technique to curb the migration and invasion of cancer cells, with a focus on the far-reaching potential of exploiting N-degron pathways for therapeutic purposes.


Sujet(s)
Autophagie/physiologie , Tumeurs/métabolisme , Proteasome endopeptidase complex/métabolisme , Protéines/métabolisme , Protéolyse , Humains
11.
Trends Biochem Sci ; 45(9): 723-725, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32616332

RÉSUMÉ

The endoplasmic reticulum-associated degradation (ERAD) pathway eliminates misfolded proteins. The Hrd1 complex represents the main gate mediating retrotranslocation of ER luminal misfolded (ERAD-L) substrates to the cytosol. A recent cryo-electron microscopy (cryo-EM) study by Wu et al. unveils the structural features of active Hrd1, providing mechanistic insights into the movement of proteins directed for degradation across ER membranes.


Sujet(s)
Cryomicroscopie électronique , Dégradation associée au réticulum endoplasmique , Réticulum endoplasmique/métabolisme , Protéolyse , Ubiquitin-protein ligases/métabolisme , Ubiquitines/métabolisme
12.
Hum Genet ; 139(12): 1513-1529, 2020 Dec.
Article de Anglais | MEDLINE | ID: mdl-32529326

RÉSUMÉ

Prader-Willi syndrome (PWS) is a neurodevelopmental disorder caused by the loss of function of a set of imprinted genes on chromosome 15q11-15q13. One of these genes, NDN, encodes necdin, a protein that is important for neuronal differentiation and survival. Loss of Ndn in mice causes defects in the formation and function of the nervous system. Necdin is a member of the melanoma-associated antigen gene (MAGE) protein family. The functions of MAGE proteins depend highly on their interactions with other proteins, and in particular MAGE proteins interact with E3 ubiquitin ligases and deubiquitinases to form MAGE-RING E3 ligase-deubiquitinase complexes. Here, we used proximity-dependent biotin identification (BioID) and mass spectrometry (MS) to determine the network of protein-protein interactions (interactome) of the necdin protein. This process yielded novel as well as known necdin-proximate proteins that cluster into a protein network. Next, we used BioID-MS to define the interactomes of necdin proteins carrying coding variants. Variant necdin proteins had interactomes that were distinct from wildtype necdin. BioID-MS is not only a useful tool to identify protein-protein interactions, but also to analyze the effects of variants of unknown significance on the interactomes of proteins involved in genetic disease.


Sujet(s)
Substitution d'acide aminé/génétique , Protéines de tissu nerveux/génétique , Protéines nucléaires/génétique , Cartes d'interactions protéiques/génétique , Ubiquitin-protein ligases/génétique , Animaux , Biotinylation/génétique , Différenciation cellulaire/génétique , Enzymes de désubiquitinylation/génétique , Régulation de l'expression des gènes/génétique , Cellules HEK293 , Humains , Spectrométrie de masse/méthodes , Souris , Mutation/génétique , Protéines de tissu nerveux/composition chimique , Protéines de tissu nerveux/ultrastructure , Malformations du système nerveux/génétique , Malformations du système nerveux/anatomopathologie , Neurones/métabolisme , Protéines nucléaires/composition chimique , Protéines nucléaires/ultrastructure , Protéines de liaison au poly(A)/composition chimique , Protéines de liaison au poly(A)/génétique , Syndrome de Prader-Willi/génétique , Conformation des protéines , Relation structure-activité , Ubiquitin-protein ligases/composition chimique
13.
Biochem Cell Biol ; 98(1): 61-69, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31095918

RÉSUMÉ

In deciphering the regulatory networks of gene expression controlled by the small non-coding RNAs known as microRNAs (miRNAs), a major challenge has been with the identification of the true mRNA targets by these RNAs within the context of the enormous numbers of predicted targets for each of these small RNAs. To facilitate the system-wide identification of miRNA targets, a variety of system wide methods, such as proteomics, have been implemented. Here we describe the utilization of quantitative label-free proteomics and bioinformatics to identify the most significant changes to the proteome upon expression of the miR-23a-27a-24-2 miRNA cluster. In light of recent work leading to the hypothesis that only the most pronounced regulatory events by miRNAs may be physiologically relevant, our data reveal that label-free analysis circumvents the limitations of proteomic labeling techniques that limit the maximum differences that can be quantified. The result of our analysis identifies a series of novel candidate targets that are reduced in abundance by more than an order of magnitude upon the expression of the miR-23a-27a-24-2 cluster.


Sujet(s)
microARN/biosynthèse , Protéome/métabolisme , Protéomique , Cellules cultivées , Cellules HEK293 , Humains , microARN/analyse , Protéome/analyse
14.
RNA ; 26(1): 44-57, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31653714

RÉSUMÉ

DDX21 is a newly discovered RNA G-quadruplex (rG4) binding protein with no known biological rG4 targets. In this study we used label-free proteomic MS/MS to identify 26 proteins that are expressed at significantly different levels in cells expressing an rG4-binding deficient DDX21 (M4). MS data are available via ProteomeXchange with identifier PXD013501. From this list we validate MAGED2 as a protein that is regulated by DDX21 through rG4 in its 5'-UTR. MAGED2 protein levels, but not mRNA levels, are reduced by half in cells expressing DDX21 M4. MAGED2 has a repressive effect on TRAIL-R2 expression that is relieved under these conditions, resulting in elevated TRAIL-R2 mRNA and protein in MCF-7 cells, rendering them sensitive to TRAIL-mediated apoptosis. Our work identifies the role of DDX21 in regulation at the translational level through biologically relevant rG4 and shows that MAGED2 protein levels are regulated, at least in part, by the potential to form rG4 in their 5'-UTRs.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Antigènes néoplasiques/métabolisme , DEAD-box RNA helicases/métabolisme , G-quadruplexes , Régulation de l'expression des gènes , ARN/génétique , Récepteurs de TRAIL/métabolisme , Régions 5' non traduites/génétique , Protéines adaptatrices de la transduction du signal/génétique , Antigènes néoplasiques/génétique , DEAD-box RNA helicases/génétique , Guanine/composition chimique , Humains , Cellules MCF-7 , Biosynthèse des protéines , Protéomique , Récepteurs de TRAIL/génétique , Spectrométrie de masse en tandem
15.
Cardiovasc Res ; 116(12): 1958-1971, 2020 10 01.
Article de Anglais | MEDLINE | ID: mdl-31742355

RÉSUMÉ

AIMS: When activated, Na+/H+ exchanger-1 (NHE1) produces some of the largest ionic fluxes in the heart. NHE1-dependent H+ extrusion and Na+ entry strongly modulate cardiac physiology through the direct effects of pH on proteins and by influencing intracellular Ca2+ handling. To attain an appropriate level of activation, cardiac NHE1 must respond to myocyte-derived cues. Among physiologically important cues is nitric oxide (NO), which regulates a myriad of cardiac functions, but its actions on NHE1 are unclear. METHODS AND RESULTS: NHE1 activity was measured using pH-sensitive cSNARF1 fluorescence after acid-loading adult ventricular myocytes by an ammonium prepulse solution manoeuvre. NO signalling was manipulated by knockout of its major constitutive synthase nNOS, adenoviral nNOS gene delivery, nNOS inhibition, and application of NO-donors. NHE1 flux was found to be activated by low [NO], but inhibited at high [NO]. These responses involved cGMP-dependent signalling, rather than S-nitros(yl)ation. Stronger cGMP signals, that can inhibit phosphodiesterase enzymes, allowed [cAMP] to rise, as demonstrated by a FRET-based sensor. Inferring from the actions of membrane-permeant analogues, cGMP was determined to activate NHE1, whereas cAMP was inhibitory, which explains the biphasic regulation by NO. Activation of NHE1-dependent Na+ influx by low [NO] also increased the frequency of spontaneous Ca2+ waves, whereas high [NO] suppressed these aberrant forms of Ca2+ signalling. CONCLUSIONS: Physiological levels of NO stimulation increase NHE1 activity, which boosts pH control during acid-disturbances and results in Na+-driven cellular Ca2+ loading. These responses are positively inotropic but also increase the likelihood of aberrant Ca2+ signals, and hence arrhythmia. Stronger NO signals inhibit NHE1, leading to a reversal of the aforementioned effects, ostensibly as a potential cardioprotective intervention to curtail NHE1 overdrive.


Sujet(s)
Myocytes cardiaques/métabolisme , Monoxyde d'azote/métabolisme , Échangeur-1 de sodium-hydrogène/métabolisme , Animaux , Signalisation calcique , Lignée cellulaire tumorale , AMP cyclique/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , GMP cyclique/métabolisme , Cyclic GMP-Dependent Protein Kinases/métabolisme , Humains , Concentration en ions d'hydrogène , Préparation de coeur isolé , Mâle , Souris knockout , Myocytes cardiaques/effets des médicaments et des substances chimiques , Donneur d'oxyde nitrique/pharmacologie , Nitric oxide synthase type I/génétique , Nitric oxide synthase type I/métabolisme , Phosphorylation , Rat Sprague-Dawley , Systèmes de seconds messagers
16.
Bioessays ; 41(11): e1800167, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31549739

RÉSUMÉ

The N-end rule denotes the relationship between the identity of the amino-terminal residue of a protein and its in vivo half-life. Since its discovery in 1986, the N-end rule has generally been described by a defined set of rules for determining whether an amino-terminal residue is stabilizing or not. However, recent studies are revealing that this N-end rule (or N-degron concept) is less straightforward than previously appreciated. For instance, it is unveiled that N-terminal acetylation of N-terminal residues may create a degradation signal (Ac-degron) that promotes the degradation of target proteins. A recent high-throughput dissection of degrons in yeast proteins amino termini intriguingly suggested that the hydrophobicity of amino-terminal residues-but not the N-terminal acetylation status-may be the indispensable feature of amino-terminal degrons. Herein, these recent advances in N-terminal acetylation and the complexity of N-terminal degradation signals in the context of the N-degron pathway are analyzed.


Sujet(s)
Protéines fongiques/métabolisme , Acétylation , Humains , Protéolyse
17.
J Neurochem ; 151(4): 520-533, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31357232

RÉSUMÉ

Protein degradation is a crucial regulatory process in maintaining cellular proteostasis. The selective degradation of intracellular proteins controls diverse cellular and biochemical processes in all kingdoms of life. Targeted protein degradation is implicated in controlling the levels of regulatory proteins as well as eliminating misfolded and any otherwise abnormal proteins. Deregulation of protein degradation is concomitant with the progression of various neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. Thus, methods of measuring metabolic half-lives of proteins greatly influence our understanding of the diverse functions of proteins in mammalian cells including neuronal cells. Historically, protein degradation rates have been studied via exploiting methods that estimate overall protein degradation or focus on few individual proteins. Notably, with the recent technical advances and developments in proteomic and imaging techniques, it is now possible to measure degradation rates of a large repertoire of defined proteins and analyze the degradation profile in a detailed spatio-temporal manner, with the aim of determining proteome-wide protein stabilities upon different physiological conditions. Herein, we discuss some of the classical and novel methods for determining protein degradation rates highlighting the crucial role of some state of art approaches in deciphering the global impact of dynamic nature of targeted degradation of cellular proteins. This article is part of the Special Issue "Proteomics".


Sujet(s)
Cellules/métabolisme , Protéolyse , Protéomique/méthodes , Homéostasie protéique , Animaux , Humains , Mammifères/métabolisme
18.
Mol Cell ; 74(4): 637-639, 2019 05 16.
Article de Anglais | MEDLINE | ID: mdl-31100244

RÉSUMÉ

Despite being among the first discovered mammalian innate immune sensor, NLRP1B (NLR pyrin domain-containing1B) activation and its molecular basis have remained elusive. Two recent studies have unveiled N-terminal degradation as a common mechanism for pathogen-mediated NLRP1B inflammasome activation in mammals.


Sujet(s)
Protéines régulatrices de l'apoptose/génétique , Immunité innée/génétique , Inflammasomes/génétique , Animaux , Humains , Inflammasomes/immunologie , Interleukine-1 bêta/génétique , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/microbiologie , Souris , Protéolyse , Cellules RAW 264.7 , Shigella flexneri/immunologie , Shigella flexneri/pathogénicité
19.
Arch Biochem Biophys ; 664: 15-23, 2019 03 30.
Article de Anglais | MEDLINE | ID: mdl-30682330

RÉSUMÉ

Isoniazid (INH) is one of the oldest drugs for the treatment of tuberculosis (TB) and is of continual clinical and research interest. The aim of the current study is to investigate the ability of INH to induce monocyte differentiation and the underlying signaling pathway involved in this phenomenon using HL-60 cells. In this study, HL-60 cells were treated with different non-cytotoxic concentrations of INH or vitamin D (a well-known inducer of monocytic differentiation) to determine key functional changes in the phenotype of these cells using several biochemical and cytobiological experiments. HL-60 cells are derived from human promyelocytic leukemia and bear some resemblance to promyelocytes, which differentiate into various cell types. INH-induced differentiation was confirmed to occur in a concentration-dependent manner through several functional markers such as nonspecific esterase activity, NADPH oxidase activity and expression of surface markers CD14 and CD16 (characteristic of monocytes). INH-induced monocytic-like differentiation in HL-60 cells and demonstrated that at least 25% of cells were differentiated within the range of the pharmacological concentrations of INH. To determine the effects of INH on HL-60 cells, we applied quantitative proteomics that revealed 32 proteins were altered significantly in pathways that could involve differentiation signals. Lastly, INH activated the ERK-1/MAPK signaling pathway based on detection of phosphorylated ERK-1. These in vitro findings in HL-60 cells warrant further study using promyelocytes or hematopoietic stem cells to evaluate the physiological capability of INH to induce monocytic differentiation that may aid in host defense against TB.


Sujet(s)
Isoniazide/pharmacologie , Monocytes/cytologie , Monocytes/effets des médicaments et des substances chimiques , Phénotype , Survie cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cellules HL-60 , Humains , Antigènes CD14/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinase 3/métabolisme , Monocytes/métabolisme , NADPH oxidase/métabolisme , Récepteurs du fragment Fc des IgG/métabolisme
20.
Trends Biochem Sci ; 44(3): 181-183, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30661830

RÉSUMÉ

Unlike prokaryotes, N-terminal formylation has been confined to a handful of mitochondrial proteins in eukaryotes. A recent study unveils a new role for eukaryotic cytoplasmic N-terminal formylation linking diverse cellular stresses to N-terminal-dependent protein degradation. These findings suggest broad cellular implications in higher eukaryotes for N-terminal methionine formylation.


Sujet(s)
Eucaryotes , Cellules eucaryotes , Méthionine , Cellules procaryotes , Protéolyse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...