Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cell Commun Signal ; 22(1): 422, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39223673

RESUMEN

Post-translational SUMOylation of nuclear and cytosolic proteins maintains homeostasis in eukaryotic cells and orchestrates programmed responses to changes in metabolic demand or extracellular stimuli. In excitable cells, SUMOylation tunes the biophysical properties and trafficking of ion channels. Ion channel SUMOylation status is determined by the opposing enzyme activities of SUMO ligases and deconjugases. Phosphorylation also plays a permissive role in SUMOylation. SUMO deconjugases have been identified for several ion channels, but their corresponding E3 ligases remain unknown. This study shows PIAS3, a.k.a. KChAP, is a bona fide SUMO E3 ligase for Kv4.2 and HCN2 channels in HEK cells, and endogenous Kv4.2 and Kv4.3 channels in cardiomyocytes. PIAS3-mediated SUMOylation at Kv4.2-K579 increases channel surface expression through a rab11a-dependent recycling mechanism. PKA phosphorylation at Kv4.2-S552 reduces the current mediated by Kv4 channels in HEK293 cells, cardiomyocytes, and neurons. This study shows PKA mediated phosphorylation blocks Kv4.2-K579 SUMOylation in HEK cells and cardiomyocytes. Together, these data identify PIAS3 as a key downstream mediator in signaling cascades that control ion channel surface expression.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico , Miocitos Cardíacos , Proteínas Inhibidoras de STAT Activados , Canales de Potasio Shal , Sumoilación , Humanos , Células HEK293 , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Animales , Miocitos Cardíacos/metabolismo , Canales de Potasio Shal/metabolismo , Canales de Potasio Shal/genética , Fosforilación , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/genética
2.
PLoS Biol ; 22(1): e3002462, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38289969

RESUMEN

Mutations in the gene encoding Cu-Zn superoxide dismutase 1 (SOD1) cause a subset of familial amyotrophic lateral sclerosis (fALS) cases. A shared effect of these mutations is that SOD1, which is normally a stable dimer, dissociates into toxic monomers that seed toxic aggregates. Considerable research effort has been devoted to developing compounds that stabilize the dimer of fALS SOD1 variants, but unfortunately, this has not yet resulted in a treatment. We hypothesized that cyclic thiosulfinate cross-linkers, which selectively target a rare, 2 cysteine-containing motif, can stabilize fALS-causing SOD1 variants in vivo. We created a library of chemically diverse cyclic thiosulfinates and determined structure-cross-linking-activity relationships. A pre-lead compound, "S-XL6," was selected based upon its cross-linking rate and drug-like properties. Co-crystallographic structure clearly establishes the binding of S-XL6 at Cys 111 bridging the monomers and stabilizing the SOD1 dimer. Biophysical studies reveal that the degree of stabilization afforded by S-XL6 (up to 24°C) is unprecedented for fALS, and to our knowledge, for any protein target of any kinetic stabilizer. Gene silencing and protein degrading therapeutic approaches require careful dose titration to balance the benefit of diminished fALS SOD1 expression with the toxic loss-of-enzymatic function. We show that S-XL6 does not share this liability because it rescues the activity of fALS SOD1 variants. No pharmacological agent has been proven to bind to SOD1 in vivo. Here, using a fALS mouse model, we demonstrate oral bioavailability; rapid engagement of SOD1G93A by S-XL6 that increases SOD1G93A's in vivo half-life; and that S-XL6 crosses the blood-brain barrier. S-XL6 demonstrated a degree of selectivity by avoiding off-target binding to plasma proteins. Taken together, our results indicate that cyclic thiosulfinate-mediated SOD1 stabilization should receive further attention as a potential therapeutic approach for fALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Animales , Ratones , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Cisteína/genética , Mutación , Superóxido Dismutasa/genética , Superóxido Dismutasa/química , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/genética
3.
Pharmacol Res ; 182: 106322, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35750299

RESUMEN

Recent studies have proposed that heteromers of µ-opioid receptors (MORs) and galanin Gal1 receptors (Gal1Rs) localized in the mesencephalon mediate the dopaminergic effects of opioids. The present study reports converging evidence, using a peptide-interfering approach combined with biophysical and biochemical techniques, including total internal reflection fluorescence microscopy, for a predominant homodimeric structure of MOR and Gal1R when expressed individually, and for their preference to form functional heterotetramers when co-expressed. Results show that a heteromerization-dependent change in the Gal1R homodimeric interface leads to a switch in G-protein coupling from inhibitory Gi to stimulatory Gs proteins. The MOR-Gal1R heterotetramer, which is thus bound to Gs via the Gal1R homodimer and Gi via the MOR homodimer, provides the framework for a canonical Gs-Gi antagonist interaction at the adenylyl cyclase level. These novel results shed light on the intense debate about the oligomeric quaternary structure of G protein-coupled receptors, their predilection for heteromer formation, and the resulting functional significance.


Asunto(s)
Analgésicos Opioides , Galanina , Analgésicos Opioides/farmacología , Mesencéfalo , Péptidos , Receptores Opioides
4.
Biomolecules ; 12(1)2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35053271

RESUMEN

The endohedral metallofullerenol Gd@C82(OH)22 has been identified as a possible antineoplastic agent that can inhibit both the growth and metastasis of cancer cells. Despite these potentially important effects, our understanding of the interactions between Gd@C82(OH)22 and biomacromolecules remains incomplete. Here, we study the interaction between Gd@C82(OH)22 and the human voltage-dependent anion channel 1 (hVDAC1), the most abundant porin embedded in the mitochondrial outer membrane (MOM), and a potential druggable target for novel anticancer therapeutics. Using in silico approaches, we observe that Gd@C82(OH)22 molecules can permeate and form stable interactions with the pore of hVDAC1. Further, this penetration can occur from either side of the MOM to elicit blockage of the pore. The binding between Gd@C82(OH)22 and hVDAC1 is largely driven by long-range electrostatic interactions. Analysis of the binding free energies indicates that it is thermodynamically more favorable for Gd@C82(OH)22 to bind to the hVDAC1 pore when it enters the channel from inside the membrane rather than from the cytoplasmic side of the protein. Multiple factors contribute to the preferential penetration, including the surface electrostatic landscape of hVDAC1 and the unique physicochemical properties of Gd@C82(OH)22. Our findings provide insights into the potential molecular interactions of macromolecular biological systems with the Gd@C82(OH)22 nanodrug.


Asunto(s)
Antineoplásicos , Fulerenos , Neoplasias , Compuestos Organometálicos , Canal Aniónico 1 Dependiente del Voltaje , Antineoplásicos/química , Antineoplásicos/farmacología , Fulerenos/química , Fulerenos/farmacología , Gadolinio/química , Gadolinio/farmacología , Humanos , Compuestos Organometálicos/farmacología , Canal Aniónico 1 Dependiente del Voltaje/antagonistas & inhibidores , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
5.
J Biol Chem ; 296: 100535, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33713702

RESUMEN

Atrial fibrillation (AF) is the most commonly diagnosed cardiac arrhythmia and is associated with increased morbidity and mortality. Currently approved AF antiarrhythmic drugs have limited efficacy and/or carry the risk of ventricular proarrhythmia. The cardiac acetylcholine activated inwardly rectifying K+ current (IKACh), composed of Kir3.1/Kir3.4 heterotetrameric and Kir3.4 homotetrameric channel subunits, is one of the best validated atrial-specific ion channels. Previous research pointed to a series of benzopyran derivatives with potential for treatment of arrhythmias, but their mechanism of action was not defined. Here, we characterize one of these compounds termed Benzopyran-G1 (BP-G1) and report that it selectively inhibits the Kir3.1 (GIRK1 or G1) subunit of the KACh channel. Homology modeling, molecular docking, and molecular dynamics simulations predicted that BP-G1 inhibits the IKACh channel by blocking the central cavity pore. We identified the unique F137 residue of Kir3.1 as the critical determinant for the IKACh-selective response to BP-G1. The compound interacts with Kir3.1 residues E141 and D173 through hydrogen bonds that proved critical for its inhibitory activity. BP-G1 effectively blocked the IKACh channel response to carbachol in an in vivo rodent model and displayed good selectivity and pharmacokinetic properties. Thus, BP-G1 is a potent and selective small-molecule inhibitor targeting Kir3.1-containing channels and is a useful tool for investigating the role of Kir3.1 heteromeric channels in vivo. The mechanism reported here could provide the molecular basis for future discovery of novel, selective IKACh channel blockers to treat atrial fibrillation with minimal side effects.


Asunto(s)
Potenciales de Acción , Antiarrítmicos/farmacología , Fibrilación Atrial/tratamiento farmacológico , Benzopiranos/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Activación del Canal Iónico , Animales , Antiarrítmicos/química , Benzopiranos/química , Humanos , Ratones , Simulación del Acoplamiento Molecular
6.
J Biol Chem ; 293(10): 3546-3561, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29317494

RESUMEN

Inwardly rectifying potassium (Kir) channels establish and regulate the resting membrane potential of excitable cells in the heart, brain, and other peripheral tissues. Phosphatidylinositol 4,5-bisphosphate (PIP2) is a key direct activator of ion channels, including Kir channels. The gasotransmitter carbon monoxide has been shown to regulate Kir channel activity by altering channel-PIP2 interactions. Here, we tested in two cellular models the effects and mechanism of action of another gasotransmitter, hydrogen sulfide (H2S), thought to play a key role in cellular responses under ischemic conditions. Direct administration of sodium hydrogen sulfide as an exogenous H2S source and expression of cystathionine γ-lyase, a key enzyme that produces endogenous H2S in specific brain tissues, resulted in comparable current inhibition of several Kir2 and Kir3 channels. This effect resulted from changes in channel-gating kinetics rather than in conductance or cell-surface localization. The extent of H2S regulation depended on the strength of the channel-PIP2 interactions. H2S regulation was attenuated when channel-PIP2 interactions were strengthened and was increased when channel-PIP2 interactions were weakened by depleting PIP2 levels. These H2S effects required specific cytoplasmic cysteine residues in Kir3.2 channels. Mutation of these residues abolished H2S inhibition, and reintroduction of specific cysteine residues back into the background of the cytoplasmic cysteine-lacking mutant rescued H2S inhibition. Molecular dynamics simulation experiments provided mechanistic insights into how potential sulfhydration of specific cysteine residues could lead to changes in channel-PIP2 interactions and channel gating.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Sulfuro de Hidrógeno/farmacología , Modelos Moleculares , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Sulfuros/farmacología , Regulación Alostérica/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Células CHO , Cricetulus , Cistationina gamma-Liasa/genética , Cistationina gamma-Liasa/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/química , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/metabolismo , Ratones , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Mutación , Oocitos/citología , Oocitos/metabolismo , Técnicas de Placa-Clamp , Fosfatidilinositol 4,5-Difosfato/química , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sulfuros/química , Sulfuros/metabolismo , Xenopus laevis
7.
J Neurosci ; 33(30): 12287-99, 2013 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-23884935

RESUMEN

Pluronic F-68, an 80% hydrophilic member of the Pluronic family of polyethylene-polypropylene-polyethylene tri-block copolymers, protects non-neuronal cells from traumatic injuries and rescues hippocampal neurons from excitotoxic and oxidative insults. F-68 interacts directly with lipid membranes and restores membrane function after direct membrane damage. Here, we demonstrate the efficacy of Pluronic F-68 in rescuing rat hippocampal neurons from apoptosis after oxygen-glucose deprivation (OGD). OGD progressively decreased neuronal survival over 48 h in a severity-dependent manner, the majority of cell death occurring after 12 h after OGD. Administration of F-68 for 48 h after OGD rescued neurons from death in a dose-dependent manner. At its optimal concentration (30 µm), F-68 rescued all neurons that would have died after the first hour after OGD. This level of rescue persisted when F-68 administration was delayed 12 h after OGD. F-68 did not alter electrophysiological parameters controlling excitability, NMDA receptor-activated currents, or NMDA-induced increases in cytosolic calcium concentrations. However, F-68 treatment prevented phosphatidylserine externalization, caspase activation, loss of mitochondrial membrane potential, and BAX translocation to mitochondria, indicating that F-68 alters apoptotic mechanisms early in the intrinsic pathway of apoptosis. The profound neuronal rescue provided by F-68 after OGD and the high level of efficacy with delayed administration indicate that Pluronic copolymers may provide a novel, membrane-targeted approach to rescuing neurons after brain ischemia. The ability of membrane-active agents to block apoptosis suggests that membranes or their lipid components play prominent roles in injury-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Hipocampo/citología , Neuronas/citología , Neuronas/efectos de los fármacos , Polietilenglicoles/farmacología , Glicoles de Propileno/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/fisiología , Caspasa 1/metabolismo , Inhibidores de Caspasas/farmacología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino , Feto/citología , Glucosa/metabolismo , Glucosa/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Mitocondrias/metabolismo , Neuronas/fisiología , Oxígeno/metabolismo , Oxígeno/farmacología , Técnicas de Placa-Clamp , Embarazo , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Proteína X Asociada a bcl-2/metabolismo
8.
Cell ; 121(1): 37-47, 2005 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-15820677

RESUMEN

Reversible, covalent modification with small ubiquitin-related modifier proteins (SUMOs) is known to mediate nuclear import/export and activity of transcription factors. Here, the SUMO pathway is shown to operate at the plasma membrane to control ion channel function. SUMO-conjugating enzyme is seen to be resident in plasma membrane, to assemble with K2P1, and to modify K2P1 lysine 274. K2P1 had not previously shown function despite mRNA expression in heart, brain, and kidney and sequence features like other two-P loop K+ leak (K2P) pores that control activity of excitable cells. Removal of the peptide adduct by SUMO protease reveals K2P1 to be a K+-selective, pH-sensitive, openly rectifying channel regulated by reversible peptide linkage.


Asunto(s)
Membrana Celular/metabolismo , Activación del Canal Iónico/fisiología , Canales de Potasio de Dominio Poro en Tándem/fisiología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Animales , Células COS , Membrana Celular/fisiología , Chlorocebus aethiops , Cisteína Endopeptidasas , Electrofisiología , Endopeptidasas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Mutación/genética , Oocitos/metabolismo , Oocitos/fisiología , Péptidos/metabolismo , Unión Proteica , Técnicas del Sistema de Dos Híbridos , Proteínas de Xenopus/metabolismo , Xenopus laevis
9.
J Neurosci ; 23(13): 5531-5, 2003 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-12843253

RESUMEN

The amyloid beta peptide (Abeta) is a product of the sequential gamma- and beta-secretase cleavage of amyloid precursor protein. Inhibitors of secretase enzymes have been proposed as a potential therapeutic strategy in the treatment of Alzheimer's disease. Here, we investigate the effect of inhibiting these key enzymes on the viability of a range of cell types. Treatment of rat cortical neurons for 24 hr with secretase inhibitors or an antibody that binds Abeta resulted in a marked reduction in cell viability, as measured by MTT reduction. Incubation with secretase inhibitors caused similar effects on other neuronal cell types (rat cerebellar granule neurons and the human SH-SY5Y cell line). Interestingly, rat astrocytes and a number of non-neuronal cell lines investigated (HEK293, DDT1-FM2, and human teratorhabdoid tumor cells) were unaffected by incubation with secretase inhibitors. The coincubation of Abeta1-40 prevented the toxicity of secretase inhibitors in neuronal cells. Abeta1-40 was protective in a concentration-dependent manner, and its effects were significant at concentrations as low at 10 pm. Importantly, the protective effects of Abeta were Abeta size-form specific, with the Abeta1-42 size form affording limited protection and the Abeta25-35 size form having very little protective effect. The present study demonstrates that inhibition of beta-or gamma-secretase activity induces death in neuronal cells. Importantly, this toxicity, which our data suggest is a consequence of a decline in neuronal Abeta levels, was absent in non-neuronal cells. This study further supports a key physiological role for the enigmatic Abeta peptide.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Neuronas/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Anticuerpos/farmacología , Ácido Aspártico Endopeptidasas , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Muerte Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Cricetinae , Endopeptidasas/efectos de los fármacos , Endopeptidasas/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Inmunohistoquímica , Riñón/citología , Riñón/efectos de los fármacos , Neocórtex/citología , Neocórtex/embriología , Neuronas/citología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar
10.
Neuroreport ; 13(12): 1553-6, 2002 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-12218704

RESUMEN

Mutations in presenilin 1 (PS1) are the major cause of autosomal dominant Alzheimer's disease. We have measured the voltage-gated K+ current in the human neuroblastoma cell line SH-SY5Y using whole-cell patch-clamp. When cells were stably transfected to over-express PS1, no change in K+ current was observed. However, over-expression of a deletion mutation (deltaE9) in PS1 led to a decreased K+ current. These changes were channel specific since no change in the Na+ current could be observed in the same cells. Confocal microscopy revealed that the K(V)3.1 K+ channel subunit had a diminished plasma membrane distribution when the deltaE9 over-expressing cells were compared to control cells. Intracellular retention of Kv3.1 is consistent with the notion that PS1 can modulate the activity and trafficking of ion channels in central neurones and implicates a compromise in electrical signalling as an underlying factor in the pathogenesis of familial Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Proteínas de la Membrana/genética , Neuroblastoma , Canales de Potasio con Entrada de Voltaje , Potasio/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Expresión Génica/fisiología , Humanos , Riñón/citología , Neuropéptidos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Presenilina-1 , Canales de Potasio Shaw , Canales de Sodio/metabolismo , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA