Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.129
Filtrer
1.
Neuroscience ; 553: 121-127, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-38992568

RÉSUMÉ

INTRODUCTION: Alzheimer's disease (AD) is characterized by accumulation of amyloid beta (Aß) and hyperphosphorylated tau (Tau-P) in the brain. Aß enhances the activity of kinases involved in the formation of Tau-P. Phosphorylation at Thr 181 determines the propagation of multiple tau phosphorylations. Aß is derived from the amyloid precursor protein (APP). Cleavage of APP by ß-secretase also initiates release of heparan sulfate (HS) from the proteoglycan glypican-1 (GPC1). OBJECTIVES: In this study, we have explored possible connections between GPC1 expression, HS release, APP processing and Tau-P formation in human neural stem cells. METHODS: GPC1 formation was suppressed by using CRISPR/Cas9 and increased by using a vector encoding GPC1. HS release from GPC1 was increased by growing cells in medium containing Arg and ascorbate. Effects were monitored by immunofluorescence microscopy and slot immunoblotting using antibodies/antisera recognizing Aß, GPC1, HS released from GPC1, total Tau, and Tau phosphorylated at Thr-181, 217 or 231. The latter have been used as blood biomarkers for AD. RESULTS: Suppression of GPC1 expression resulted in increased phosphorylation at Thr 181 and Thr 217. When GPC1 was overexpressed, phosphorylation at Thr 217 decreased. Stimulation of HS release from GPC1 diminished tau phosphorylation at all of the three Thr positions, while expression of GPC1 was unaffected. Simultaneous stimulation of HS release and APP processing by the cytokine TNF-α also suppressed tau phosphorylation. CONCLUSION: The increased release of GPC1-derived HS may interfere with Aß formation and/or Aß interaction with tau.


Sujet(s)
Peptides bêta-amyloïdes , Glypicanes , Cellules souches neurales , Protéines tau , Humains , Protéines tau/métabolisme , Glypicanes/métabolisme , Phosphorylation/physiologie , Cellules souches neurales/métabolisme , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/métabolisme
2.
Dev Neurobiol ; 84(3): 217-235, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38837880

RÉSUMÉ

The Pcdhg gene cluster encodes 22 γ-Protocadherin (γ-Pcdh) cell adhesion molecules that critically regulate multiple aspects of neural development, including neuronal survival, dendritic and axonal arborization, and synapse formation and maturation. Each γ-Pcdh isoform has unique protein domains-a homophilically interacting extracellular domain and a juxtamembrane cytoplasmic domain-as well as a C-terminal cytoplasmic domain shared by all isoforms. The extent to which isoform-specific versus shared domains regulate distinct γ-Pcdh functions remains incompletely understood. Our previous in vitro studies identified protein kinase C (PKC) phosphorylation of a serine residue within a shared C-terminal motif as a mechanism through which γ-Pcdh promotion of dendrite arborization via myristoylated alanine-rich C-kinase substrate (MARCKS) is abrogated. Here, we used CRISPR/Cas9 genome editing to generate two new mouse lines expressing only non-phosphorylatable γ-Pcdhs, due either to a serine-to-alanine mutation (PcdhgS/A) or to a 15-amino acid C-terminal deletion resulting from insertion of an early stop codon (PcdhgCTD). Both lines are viable and fertile, and the density and maturation of dendritic spines remain unchanged in both PcdhgS/A and PcdhgCTD cortex. Dendrite arborization of cortical pyramidal neurons, however, is significantly increased in both lines, as are levels of active MARCKS. Intriguingly, despite having significantly reduced levels of γ-Pcdh proteins, the PcdhgCTD mutation yields the strongest phenotype, with even heterozygous mutants exhibiting increased arborization. The present study confirms that phosphorylation of a shared C-terminal motif is a key γ-Pcdh negative regulation point and contributes to a converging understanding of γ-Pcdh family function in which distinct roles are played by both individual isoforms and discrete protein domains.


Sujet(s)
Protéines apparentées aux cadhérines , Cadhérines , Cortex cérébral , Dendrites , Protéine kinase C , Animaux , Cortex cérébral/métabolisme , Cortex cérébral/cytologie , Cadhérines/métabolisme , Cadhérines/génétique , Phosphorylation/physiologie , Dendrites/métabolisme , Souris , Protéine kinase C/métabolisme , Protéine kinase C/génétique , Protéine myristoylée riche en alanine et substrat de la kinase C/métabolisme , Protéine myristoylée riche en alanine et substrat de la kinase C/génétique , Motifs d'acides aminés/physiologie , Souris transgéniques
3.
Trends Neurosci ; 47(7): 480-490, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38862330

RÉSUMÉ

α-Synuclein (αS) is an abundant presynaptic protein that regulates neurotransmission. It is also a key protein implicated in a broad class of neurodegenerative disorders termed synucleinopathies, including Parkinson's disease (PD) and Lewy body dementia (LBD). Pathological αS deposits in these diseases, Lewy bodies (LBs)/neurites (LNs), contain about 90% of αS in its phospho-serine129 (pS129) form. Therefore, pS129 is widely used as a surrogate marker of pathology. However, recent findings demonstrate that pS129 is also physiologically triggered by neuronal activity to positively regulate synaptic transmission. In this opinion article, we contrast the literature on pathological and physiological pS129, with a special focus on the latter. We emphasize that pS129 is ambiguous and knowledge about the context is necessary to correctly interpret changes in pS129.


Sujet(s)
alpha-Synucléine , alpha-Synucléine/métabolisme , Humains , Phosphorylation/physiologie , Animaux , Sérine/métabolisme , Maladie de Parkinson/métabolisme , Transmission synaptique/physiologie , Synucléinopathies/métabolisme
4.
eNeuro ; 11(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38918052

RÉSUMÉ

The zebrafish, a widely used model in neurobiology, relies on hearing in aquatic environments. Unfortunately, its auditory pathways have mainly been studied in larvae. In this study, we examined the involvement of the anterior tuberal nucleus (AT) in auditory processing in adult zebrafish. Our tract-tracing experiments revealed that the dorsal subdivision of AT is strongly bidirectionally connected to the central nucleus of the torus semicircularis (TSc), a major auditory nucleus in fishes. Immunohistochemical visualization of the ribosomal protein S6 (pS6) phosphorylation to map neural activity in response to auditory stimulation substantiated this finding: the dorsal but not the ventral part of AT responded strongly to auditory stimulation. A similar response to auditory stimulation was present in the TSc but not in the nucleus isthmi, a visual region, which we used as a control for testing if the pS6 activation was specific to the auditory stimulation. We also measured the time course of pS6 phosphorylation, which was previously unreported in teleost fish. After auditory stimulation, we found that pS6 phosphorylation peaked between 100 and 130 min and returned to baseline levels after 190 min. This information will be valuable for the design of future pS6 experiments. Our results suggest an anatomical and functional subdivision of AT, where only the dorsal part connects to the auditory network and processes auditory information.


Sujet(s)
Stimulation acoustique , Voies auditives , Danio zébré , Animaux , Danio zébré/physiologie , Voies auditives/physiologie , Phosphorylation/physiologie , Protéine ribosomique S6/métabolisme , Perception auditive/physiologie , Techniques de traçage neuroanatomique , Mâle , Femelle
5.
ACS Chem Neurosci ; 15(12): 2408-2419, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38752226

RÉSUMÉ

Huntington's disease is a neurodegenerative disorder caused by an expanded polyglutamine stretch near the N-terminus of the huntingtin (HTT) protein, rendering the protein more prone to aggregate. The first 17 residues in HTT (Nt17) interact with lipid membranes and harbor multiple post-translational modifications (PTMs) that can modulate HTT conformation and aggregation. In this study, we used a combination of biophysical studies and molecular simulations to investigate the effect of PTMs on the helicity of Nt17 in the presence of various lipid membranes. We demonstrate that anionic lipids such as PI4P, PI(4,5)P2, and GM1 significantly enhance the helical structure of unmodified Nt17. This effect is attenuated by single acetylation events at K6, K9, or K15, whereas tri-acetylation at these sites abolishes Nt17-membrane interaction. Similarly, single phosphorylation at S13 and S16 decreased but did not abolish the POPG and PIP2-induced helicity, while dual phosphorylation at these sites markedly diminished Nt17 helicity, regardless of lipid composition. The helicity of Nt17 with phosphorylation at T3 is insensitive to the membrane environment. Oxidation at M8 variably affects membrane-induced helicity, highlighting a lipid-dependent modulation of the Nt17 structure. Altogether, our findings reveal differential effects of PTMs and crosstalks between PTMs on membrane interaction and conformation of HTT. Intriguingly, the effects of phosphorylation at T3 or single acetylation at K6, K9, and K15 on Nt17 conformation in the presence of certain membranes do not mirror that observed in the absence of membranes. Our studies provide novel insights into the complex relationship between Nt17 structure, PTMs, and membrane binding.


Sujet(s)
Protéine huntingtine , Maturation post-traductionnelle des protéines , Maturation post-traductionnelle des protéines/physiologie , Protéine huntingtine/métabolisme , Protéine huntingtine/génétique , Humains , Phosphorylation/physiologie , Acétylation , Membrane cellulaire/métabolisme , Simulation de dynamique moléculaire , Lipides membranaires/métabolisme , Maladie de Huntington/métabolisme
6.
Am J Pathol ; 194(5): 785-795, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38311118

RÉSUMÉ

Necroptosis, considered as a form of programmed cell death, contributes to neural loss. The 5-hydroxytryptamine 4 receptor (5-HT4R) is involved in neurogenesis in the enteric nervous system. However, whether the activation of 5-HT4R can alleviate diabetic enteric neuropathy by inhibiting receptor-interacting protein kinase 3 (RIPK3)-mediated necroptosis is unclear. This study aimed to explore the beneficial effects of 5-HT4R agonist on enteric neuropathy in a mouse model of diabetes and the mechanisms underlying these effects. Diabetes developed neural loss in the colon of mice. 5-HT4Rs localized in submucosal and myenteric plexuses were confirmed. Administration of 5-HT4R agonist attenuated diabetes-induced colonic hypomotility and neural loss of the colon in mice. Remarkably, RIPK3, phosphorylated RIPK3, and its downstream target mixed lineage kinase domain-like protein (MLKL), two key proteins regulating necroptosis, were significantly up-regulated in the colon of diabetic mice. Treatment with 5-HT4R agonist appeared to inhibit diabetes-induced elevation of RIPK3, phosphorylated RIPK3, and MLKL in the colon of mice. Diabetes-induced up-regulation of MLKL in both the mucosa and the muscularis of the colon was prevented by Ripk3 deletion. Moreover, diabetes-evoked neural loss and delayed colonic transit were significantly inhibited by Ripk3 removal. These findings suggest that activation of 5-HT4Rs could potentially provide a protective effect against diabetic enteric neuropathy by suppressing RIPK3-mediated necroptosis.


Sujet(s)
Diabète expérimental , Protein kinases , Souris , Animaux , Protein kinases/métabolisme , Sérotonine/métabolisme , Diabète expérimental/complications , Diabète expérimental/traitement médicamenteux , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Apoptose , Phosphorylation/physiologie
7.
Brain Res ; 1829: 148793, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38309553

RÉSUMÉ

Alzheimer's disease (AD) is a progressive neurological disorder that impairs memory and cognitive abilities, primarily in the elderly. The burden of AD extends beyond patients, impacting families and caregivers due to the patients' reliance on assistance for daily tasks. The main features of the pathogenesis of AD are beta-amyloid plaques and neurofibrillary tangles (NFTs), that strongly correlate with oxidative stress and inflammation. NFTs result from misfolded and hyperphosphorylated tau proteins. Various studies have focused on tau phosphorylation, indicating protein phosphatase 2A (PP2A) as the primary tau phosphatase and glycogen synthase kinase-3 beta (GSK-3ß) as the leading tau kinase. Experimental evidence suggests that inhibition of PP2A and increased GSK-3ß activity contribute to neuroinflammation, oxidative stress, and cognitive impairment. Hence, targeting PP2A and GSK-3ß with pharmacological approaches shows promise in treating AD. The use of natural compounds in the drug development for AD have been extensively studied for their antioxidant, anti-inflammatory, anti-cholinesterase, and neuroprotective properties, demonstrating therapeutic advantages in neurological diseases. Alongside the development of PP2A activator and GSK-3ß inhibitor drugs, natural compounds are likely to have neuroprotective effects by increasing PP2A activity and decreasing GSK-3ß levels. Therefore, based on the preclinical and clinical studies, the potential of PP2A and GSK-3ß as therapeutic targets of natural compounds are highlighted in this review.


Sujet(s)
Maladie d'Alzheimer , Humains , Sujet âgé , Maladie d'Alzheimer/métabolisme , Protein Phosphatase 2/métabolisme , Glycogen synthase kinase 3 beta/métabolisme , Protéines tau/métabolisme , Peptides bêta-amyloïdes/métabolisme , Phosphorylation/physiologie
8.
Sci Adv ; 10(6): eadj2752, 2024 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-38324677

RÉSUMÉ

Exercise-induced activation of adenosine monophosphate-activated protein kinase (AMPK) and substrate phosphorylation modulate the metabolic capacity of mitochondria in skeletal muscle. However, the key effector(s) of AMPK and the regulatory mechanisms remain unclear. Here, we showed that AMPK phosphorylation of the folliculin interacting protein 1 (FNIP1) serine-220 (S220) controls mitochondrial function and muscle fuel utilization during exercise. Loss of FNIP1 in skeletal muscle resulted in increased mitochondrial content and augmented metabolic capacity, leading to enhanced exercise endurance in mice. Using skeletal muscle-specific nonphosphorylatable FNIP1 (S220A) and phosphomimic (S220D) transgenic mouse models as well as biochemical analysis in primary skeletal muscle cells, we demonstrated that exercise-induced FNIP1 (S220) phosphorylation by AMPK in muscle regulates mitochondrial electron transfer chain complex assembly, fuel utilization, and exercise performance without affecting mechanistic target of rapamycin complex 1-transcription factor EB signaling. Therefore, FNIP1 is a multifunctional AMPK effector for mitochondrial adaptation to exercise, implicating a mechanism for exercise tolerance in health and disease.


Sujet(s)
AMP-Activated Protein Kinases , Protéines de transport , Souris , Animaux , Phosphorylation/physiologie , AMP-Activated Protein Kinases/métabolisme , Protéines de transport/métabolisme , Mitochondries/métabolisme , Muscles squelettiques/métabolisme
9.
Exp Mol Med ; 56(1): 210-219, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38200156

RÉSUMÉ

Traumatic brain injury (TBI) is a major cause of death and disability in adults. The pathological process of TBI involves a multifactorial cascade in which kinases have been proven contribute to interactions between relevant factors and amplification of signaling cascades. Cyclin-dependent kinase 5 (Cdk5) is a promising kinase that has been implicated in various brain disorders, including TBI. However, the mechanism by which Cdk5 induces neuronal damage remains unclear. Here, we show for the first time that Drosha, a key enzyme in microRNA biogenesis, is a pivotal substrate of abnormally activated Cdk5. Cdk5-mediated phosphorylation decreases Drosha expression and exacerbates nerve injury in TBI. We proved that maintaining Drosha expression via the administration of repurposed Cdk5 inhibitors that were previously studied in clinical trials is a promising approach for the early treatment of TBI. Together, our work identifies Drosha as a novel target for neuroprotective strategies after TBI and suggests Cdk5-mediated regulation of Drosha expression as a potential therapeutic strategy for early TBI intervention.


Sujet(s)
Lésions traumatiques de l'encéphale , Humains , Phosphorylation/physiologie , Lésions traumatiques de l'encéphale/traitement médicamenteux , Lésions traumatiques de l'encéphale/métabolisme , Transduction du signal/physiologie , Kinase-5 cycline-dépendante/génétique , Kinase-5 cycline-dépendante/métabolisme
10.
J Mol Biol ; 436(2): 168379, 2024 01 15.
Article de Anglais | MEDLINE | ID: mdl-38043732

RÉSUMÉ

Phosphosignaling in bacteria is mediated by two distinct systems, the two-component systems (TCSs) and the protein Ser/Thr/Tyr, or O-phosphorylation systems. These two arms of phosphosignaling are currently thought to be largely independent from one another. We mined a deep Mycobacterium tuberculosis (Mtb) phosphoproteome and identified over 170 O-phosphorylation sites on histidine kinases and response regulators of TCSs, suggesting that the two signaling pathways extensively intersect. Several TCSs were phosphorylated on multiple sites, and many by multiple Ser/Thr protein kinases, suggesting convergent and cooperative regulatory interactions. To test in which way these O-phosphorylation sites affect TCS activity, we reconstituted the NarSL phosphorelay in vitro. The Ser/Thr protein kinase PknL phosphorylated the histidine kinase NarS and activated its autophosphorylating activity. A phosphoablative mutation at the PknL phosphorylation site Thr380 resulted in low autophosphorylating activity, whereas a phosphomimetic mutation strongly activated autophosphorylation. The phosphomimetic mutation also resulted in more efficient phosphotransfer from NarS to the response regulator NarL and suppression of gene expression. These data show control of NarSL signaling by STPKs through a phosphoswitch and point to extensive, functional crosstalk between TCSs and O-phosphorylation.


Sujet(s)
Protéines bactériennes , Mycobacterium tuberculosis , Protein-Serine-Threonine Kinases , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Mycobacterium tuberculosis/enzymologie , Mycobacterium tuberculosis/génétique , Phosphorylation/physiologie , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme
11.
Nat Commun ; 14(1): 7920, 2023 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-38040726

RÉSUMÉ

Many functional aspects of the protein kinase p38α have been illustrated by more than three hundred structures determined in the presence of reducing agents. These structures correspond to free forms and complexes with activators, substrates, and inhibitors. Here we report the conformation of an oxidized state with an intramolecular disulfide bond between Cys119 and Cys162 that is conserved in vertebrates. The structure of the oxidized state does not affect the conformation of the catalytic site, but alters the docking groove by partially unwinding and displacing the short αD helix due to the movement of Cys119 towards Cys162. The transition between oxidized and reduced conformations provides a mechanism for fine-tuning p38α activity as a function of redox conditions, beyond its activation loop phosphorylation. Moreover, the conformational equilibrium between these redox forms reveals an unexplored cleft for p38α inhibitor design that we describe in detail.


Sujet(s)
Mitogen-Activated Protein Kinase 14 , Animaux , Conformation des protéines , Mitogen-Activated Protein Kinase 14/métabolisme , Phosphorylation/physiologie , Domaine catalytique , Oxydoréduction
12.
Sci Rep ; 13(1): 21660, 2023 12 08.
Article de Anglais | MEDLINE | ID: mdl-38066086

RÉSUMÉ

The potassium chloride cotransporter KCC2 is crucial for Cl- extrusion from mature neurons and thus key to hyperpolarizing inhibition. Auditory brainstem circuits contain well-understood inhibitory projections and provide a potent model to study the regulation of synaptic inhibition. Two peculiarities of the auditory brainstem are (i) posttranslational activation of KCC2 during development and (ii) extremely negative reversal potentials in specific circuits. To investigate the role of the potent phospho-site serine 937 therein, we generated a KCC2 Thr934Ala/Ser937Asp double mutation, in which Ser937 is replaced by aspartate mimicking the phosphorylated state, and the neighbouring Thr934 arrested in the dephosphorylated state. This double mutant showed a twofold increased transport activity in HEK293 cells, raising the hypothesis that auditory brainstem neurons show lower [Cl-]i. and increased glycinergic inhibition. This was tested in a mouse model carrying the same KCC2 Thr934Ala/Ser937Asp mutation by the use of the CRISPR/Cas9 technology. Homozygous KCC2 Thr934Ala/Ser937Asp mice showed an earlier developmental onset of hyperpolarisation in the auditory brainstem. Mature neurons displayed stronger glycinergic inhibition due to hyperpolarized ECl-. These data demonstrate that phospho-regulation of KCC2 Ser937 is a potent way to interfere with the excitation-inhibition balance in neural circuits.


Sujet(s)
, Sérine , Animaux , Humains , Souris , Cellules HEK293 , /métabolisme , Neurones/métabolisme , Phosphorylation/physiologie , Sérine/métabolisme
14.
Sci Adv ; 9(46): eadh1110, 2023 11 17.
Article de Anglais | MEDLINE | ID: mdl-37967196

RÉSUMÉ

Synaptic plasticity plays a crucial role in memory formation by regulating the communication between neurons. Although actin polymerization has been linked to synaptic plasticity and dendritic spine stability, the causal link between actin polymerization and memory encoding has not been identified yet. It is not clear whether actin polymerization and structural changes in dendritic spines are a driver or a consequence of learning and memory. Using an extrinsically disordered form of the protein kinase LIMK1, which rapidly and precisely acts on ADF/cofilin, a direct modifier of actin, we induced long-term enlargement of dendritic spines and enhancement of synaptic transmission in the hippocampus on command. The activation of extrinsically disordered LIMK1 in vivo improved memory encoding and slowed cognitive decline in aged mice exhibiting reduced cofilin phosphorylation. The engineered memory by an extrinsically disordered LIMK1 supports a direct causal link between actin-mediated synaptic transmission and memory.


Sujet(s)
Actines , Hippocampe , Souris , Animaux , Actines/métabolisme , Hippocampe/métabolisme , Facteurs de dépolymérisation de l'actine/métabolisme , Phosphorylation/physiologie , Plasticité neuronale/physiologie
15.
Cardiovasc Diabetol ; 22(1): 276, 2023 10 13.
Article de Anglais | MEDLINE | ID: mdl-37833717

RÉSUMÉ

BACKGROUND: O-GlcNAcylation is the enzymatic addition of a sugar, O-linked ß-N-Acetylglucosamine, to the serine and threonine residues of proteins, and is abundant in diabetic conditions. We have previously shown that O-GlcNAcylation can trigger arrhythmias by indirectly increasing pathological Ca2+ leak through the cardiac ryanodine receptor (RyR2) via Ca2+/calmodulin-dependent kinase II (CaMKII). However, RyR2 is well known to be directly regulated by other forms of serine and threonine modification, therefore, this study aimed to determine whether RyR2 is directly modified by O-GlcNAcylation and if this also alters the function of RyR2 and Ca2+ leak. METHODS: O-GlcNAcylation of RyR2 in diabetic human and animal hearts was determined using western blotting. O-GlcNAcylation of RyR2 was pharmacologically controlled and the propensity for Ca2+ leak was determined using single cell imaging. The site of O-GlcNAcylation within RyR2 was determined using site-directed mutagenesis of RyR2. RESULTS: We found that RyR2 is modified by O-GlcNAcylation in human, animal and HEK293 cell models. Under hyperglycaemic conditions O-GlcNAcylation was associated with an increase in Ca2+ leak through RyR2 which persisted after CaMKII inhibition. Conversion of serine-2808 to alanine prevented an O-GlcNAcylation induced increase in Ca2+ leak. CONCLUSIONS: These data suggest that the function of RyR2 can be directly regulated by O-GlcNAcylation and requires the presence of serine-2808.


Sujet(s)
Diabète , Canal de libération du calcium du récepteur à la ryanodine , Animaux , Humains , Canal de libération du calcium du récepteur à la ryanodine/génétique , Canal de libération du calcium du récepteur à la ryanodine/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Myocytes cardiaques/métabolisme , Cellules HEK293 , Phosphorylation/physiologie , Réticulum sarcoplasmique/métabolisme , Diabète/métabolisme , Sérine/métabolisme , Thréonine/métabolisme , Calcium/métabolisme
16.
J Neurosci ; 43(44): 7264-7275, 2023 11 01.
Article de Anglais | MEDLINE | ID: mdl-37699715

RÉSUMÉ

Neuroligins (NLGNs) are postsynaptic cell adhesion molecules that are involved in synapse assembly and function. The NLGN gene family consists of 5 genes (NLGN1-3, 4X, and 4Y). NLGN3 forms heterodimers with other NLGNs and is expressed at both excitatory and inhibitory synapses, although the distinct role at different synapses is not fully understood. Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase that targets various neuronal substrates to impact neuronal migration, neurite outgrowth, synaptic transmission, and plasticity. Both NLGNs and their presynaptic binding partners neurexins are highly associated with neurodevelopmental disorders. The NLGN3 gene is on the X chromosome and variants in NLGN3 have been linked to the pathophysiology in neurodevelopmental disorders. To better understand the endogenous modulation of NLGN3, we generated an HA-tagged knock-in mouse. We found that Cdk5 associates with NLGN3 in vivo and phosphorylates NLGN3 on serine 725 (S725) in the knock-in mouse of either sex. The phosphorylation affects the NLGN3 association with Kalirin-7, a postsynaptic guanine nucleotide exchange factors for Rho GTPase family proteins. We further observed that the phosphorylation modulates NLGN3 surface expression and NLGN3-mediated synaptic currents in cultured rat neurons. Thus, we characterized NLGN3 as a novel Cdk5 substrate and revealed the functional consequences of NLGN3 S725 phosphorylation in neurons. Our study provides a novel molecular mechanism underlying Cdk5-mediated regulation of postsynaptic cell adhesion molecules.SIGNIFICANCE STATEMENT NLGN3 is involved in synapse assembly and function at both excitatory and inhibitory synapses and has been associated with the pathophysiology of neurodevelopmental disorders. Cdk5 has brain-specific activity and is involved in neuronal transmission, synapse function, and plasticity. Here, we characterize NLGN3 as a Cdk5 substrate for the first time and show that Cdk5-mediated phosphorylation regulates NLGN3 function. We demonstrate that NLGN3 S725 is a Cdk5 phosphorylation site, and reveal that the site is important for NLGN3 association with Kalirin-7, NLGN3 surface expression, and NLGN3-mediated synaptic transmission.


Sujet(s)
Kinase-5 cycline-dépendante , Synapses , Animaux , Souris , Rats , Molécules d'adhérence cellulaire/métabolisme , Kinase-5 cycline-dépendante/génétique , Kinase-5 cycline-dépendante/métabolisme , Phosphorylation/physiologie , Rho guanine nucleotide exchange factors/métabolisme , Sérine/métabolisme , Synapses/métabolisme , Transmission synaptique
17.
Cell Death Dis ; 14(7): 436, 2023 07 15.
Article de Anglais | MEDLINE | ID: mdl-37454104

RÉSUMÉ

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common genetic cause of Parkinson's disease (PD), with growing importance also for Crohn's disease and cancer. LRRK2 is a large and complex protein possessing both GTPase and kinase activity. Moreover, LRRK2 activity and function can be influenced by its phosphorylation status. In this regard, many LRRK2 PD-associated mutants display decreased phosphorylation of the constitutive phosphorylation cluster S910/S935/S955/S973, but the role of these changes in phosphorylation status with respect to LRRK2 physiological functions remains unknown. Here, we propose that the S910/S935/S955/S973 phosphorylation sites act as key regulators of LRRK2-mediated autophagy under both basal and starvation conditions. We show that quadruple LRRK2 phosphomutant cells (4xSA; S910A/S935A/S955A/S973A) have impaired lysosomal functionality and fail to induce and proceed with autophagy during starvation. In contrast, treatment with the specific LRRK2 kinase inhibitors MLi-2 (100 nM) or PF-06447475 (150 nM), which also led to decreased LRRK2 phosphorylation of S910/S935/S955/S973, did not affect autophagy. In explanation, we demonstrate that the autophagy impairment due to the 4xSA LRRK2 phospho-dead mutant is driven by its enhanced LRRK2 kinase activity. We show mechanistically that this involves increased phosphorylation of LRRK2 downstream targets Rab8a and Rab10, as the autophagy impairment in 4xSA LRRK2 cells is counteracted by expression of phosphorylation-deficient mutants T72A Rab8a and T73A Rab10. Similarly, reduced autophagy and decreased LRRK2 phosphorylation at the constitutive sites were observed in cells expressing the pathological R1441C LRRK2 PD mutant, which also displays increased kinase activity. These data underscore the relation between LRRK2 phosphorylation at its constitutive sites and the importance of increased LRRK2 kinase activity in autophagy regulation and PD pathology.


Sujet(s)
Autophagie , Protéines G rab , Phosphorylation/physiologie , Leucine-rich repeat serine-threonine protein kinase-2/génétique , Leucine-rich repeat serine-threonine protein kinase-2/métabolisme , Mutation , Autophagie/génétique , Protéines G rab/génétique , Protéines G rab/métabolisme
18.
Cell Rep ; 42(7): 112729, 2023 07 25.
Article de Anglais | MEDLINE | ID: mdl-37405922

RÉSUMÉ

Protein phosphorylation modification is crucial for signaling transduction in plant development and environmental adaptation. By precisely phosphorylating crucial components in signaling cascades, plants can switch on and off the specific signaling pathways needed for growth or defense. Here, we have summarized recent findings of key phosphorylation events in typical hormone signaling and stress responses. More interestingly, distinct phosphorylation patterns on proteins result in diverse biological functions of these proteins. Thus, we have also highlighted latest findings that show how the different phosphosites of a protein, also named phosphocodes, determine the specificity of downstream signaling in both plant development and stress responses.


Sujet(s)
Plantes , Transduction du signal , Phosphorylation/physiologie , Développement des plantes , Protéines végétales/métabolisme
19.
Cells ; 12(12)2023 06 06.
Article de Anglais | MEDLINE | ID: mdl-37371033

RÉSUMÉ

Arrestins bind active phosphorylated G protein-coupled receptors (GPCRs). Among the four mammalian subtypes, only arrestin-3 facilitates the activation of JNK3 in cells. In available structures, Lys-295 in the lariat loop of arrestin-3 and its homologue Lys-294 in arrestin-2 directly interact with the activator-attached phosphates. We compared the roles of arrestin-3 conformational equilibrium and Lys-295 in GPCR binding and JNK3 activation. Several mutants with enhanced ability to bind GPCRs showed much lower activity towards JNK3, whereas a mutant that does not bind GPCRs was more active. The subcellular distribution of mutants did not correlate with GPCR recruitment or JNK3 activation. Charge neutralization and reversal mutations of Lys-295 differentially affected receptor binding on different backgrounds but had virtually no effect on JNK3 activation. Thus, GPCR binding and arrestin-3-assisted JNK3 activation have distinct structural requirements, suggesting that facilitation of JNK3 activation is the function of arrestin-3 that is not bound to a GPCR.


Sujet(s)
Arrestines , Récepteurs couplés aux protéines G , Animaux , bêta-Arrestine 2/métabolisme , Phosphorylation/physiologie , Arrestines/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Liaison aux protéines/physiologie , Mammifères/métabolisme
20.
Int J Mol Sci ; 24(12)2023 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-37373182

RÉSUMÉ

A class-A GPCR dopamine D2 receptor (D2R) plays a critical role in the proper functioning of neuronal circuits through the downstream activation of both G-protein- and ß-arrestin-dependent signaling pathways. Understanding the signaling pathways downstream of D2R is critical for developing effective therapies with which to treat dopamine (DA)-related disorders such as Parkinson's disease and schizophrenia. Extensive studies have focused on the regulation of D2R-mediated extracellular-signal-regulated kinase (ERK) 1/2 signaling; however, the manner in which ERKs are activated upon the stimulation of a specific signaling pathway of D2R remains unclear. The present study conducted a variety of experimental techniques, including loss-of-function experiments, site-directed mutagenesis, and the determination of protein interactions, in order to investigate the mechanisms underlying ß-arrestin-biased signaling-pathway-mediated ERK activation. We found that the stimulation of the D2R ß-arrestin signaling pathway caused Mdm2, an E3 ubiquitin ligase, to move from the nucleus to the cytoplasm and interact with tyrosine phosphorylated G-protein-coupled receptor kinase 2 (GRK2), which was facilitated by Src, a non-receptor tyrosine kinase. This interaction led to the ubiquitination of GRK2, which then moved to the plasma membrane and interacted with activated D2R, followed by the phosphorylation of D2R as well as the mediation of ERK activation. In conclusion, Mdm2-mediated GRK2 ubiquitination, which is selectively triggered by the stimulation of the D2R ß-arrestin signaling pathway, is necessary for GRK2 membrane translocation and its interaction with D2R, which in turn mediates downstream ERK signaling. This study is primarily novel and provides essential information with which to better understand the detailed mechanisms of D2R-dependent signaling.


Sujet(s)
Kinase-2 associée au récepteur couplé à une protéine G , Transduction du signal , bêta-Arrestines/métabolisme , Kinase-2 associée au récepteur couplé à une protéine G/génétique , Kinase-2 associée au récepteur couplé à une protéine G/métabolisme , Transduction du signal/physiologie , bêta-Arrestine 1/métabolisme , Récepteur D2 de la dopamine/génétique , Récepteur D2 de la dopamine/métabolisme , Phosphorylation/physiologie , Dopamine , Ubiquitination
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE