Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35.367
Filtrar
1.
Commun Biol ; 7(1): 1264, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39367191

RESUMEN

The negative membrane potential within bacterial cells is crucial in various essential cellular processes. Sustaining a hyperpolarised membrane could offer a novel strategy to combat antimicrobial resistance. However, it remains uncertain which molecules are responsible for inducing hyperpolarization and what the underlying molecular mechanisms are. Here, we demonstrate that chemically diverse antimicrobial peptides (AMPs) trigger hyperpolarization of the bacterial cytosolic membrane when applied at subinhibitory concentrations. Specifically, these AMPs adopt a membrane-induced amphipathic structure and, thereby, generate hyperpolarization in Escherichia coli without damaging the cell membrane. These AMPs act as selective ionophores for K+ (over Na+) or Cl- (over H2PO4- and NO3-) ions, generating diffusion potential across the membrane. At lower dosages of AMPs, a quasi-steady-state membrane polarisation value is achieved. Our findings highlight the potential of AMPs as a valuable tool for chemically hyperpolarising bacteria, with implications for antimicrobial research and bacterial electrophysiology.


Asunto(s)
Péptidos Antimicrobianos , Membrana Celular , Escherichia coli , Potenciales de la Membrana , Escherichia coli/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Antibacterianos/farmacología , Antibacterianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/química
2.
Sci Rep ; 14(1): 24940, 2024 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-39438518

RESUMEN

Constrictor agonists set arterial tone through two coupling processes, one tied to (electromechanical), the other independent (pharmacomechanical) of, membrane potential (VM). This dual arrangement raises an intriguing question: is the contribution of each mechanism (1) fixed and proportionate, or (2) variable and functionally biased. Examination began in mouse mesenteric arteries with a vasomotor assessment to a classic Gq/11 (phenylephrine) or Gq/11/G12/13 (U46619) agonist, in the absence and presence of nifedipine, to separate among the two coupling mechanisms. Each constrictor elicited a concentration response curve that was attenuated and rightward shifted by nifedipine, findings consistent with functional bias. Electromechanical coupling preceded pharmacomechanical, the latter's importance rising with agonist concentration. In this regard, ensuing contractile and phosphorylation (CPI-17 & MYPT1 (T-855 & T-697)) measures revealed phenylephrine-induced pharmacomechanical coupling was tied to protein kinase C (PKC) activity, while that enabled by U46619 to PKC and Rho-kinase. A complete switch to pharmacomechanical coupling arose when agonist superfusion was replaced by pipet application to a small portion of artery. This switch was predicted, a priori, by a computer model of electromechanical control and supported by additional measures of VM and cytosolic Ca2+. We conclude that the coupling mechanisms driving agonist-induced constriction are variable and functionally biased, their relative importance set in accordance with agonist concentration and manner of application. These findings have important implications to hemodynamic control in health and disease, including hypertension and arterial vasospasm.


Asunto(s)
Arterias Mesentéricas , Fenilefrina , Vasoconstricción , Vasoconstrictores , Animales , Ratones , Vasoconstricción/efectos de los fármacos , Fenilefrina/farmacología , Arterias Mesentéricas/fisiología , Arterias Mesentéricas/efectos de los fármacos , Vasoconstrictores/farmacología , Masculino , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Proteína Quinasa C/metabolismo , Nifedipino/farmacología , Fosforilación , Resistencia Vascular/efectos de los fármacos , Ratones Endogámicos C57BL , Potenciales de la Membrana/efectos de los fármacos
3.
Crit Care Med ; 52(10): 1499-1508, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39312458

RESUMEN

OBJECTIVES: Hyperkalemia is a common life-threatening condition causing severe electrophysiologic derangements and arrhythmias. The beneficial effects of calcium (Ca 2+ ) treatment for hyperkalemia have been attributed to "membrane stabilization," by restoration of resting membrane potential (RMP). However, the underlying mechanisms remain poorly understood. Our objective was to investigate the mechanisms underlying adverse electrophysiologic effects of hyperkalemia and the therapeutic effects of Ca 2+ treatment. DESIGN: Controlled experimental trial. SETTING: Laboratory investigation. SUBJECTS: Canine myocytes and tissue preparations. INTERVENTIONS AND MEASUREMENTS: Optical action potentials and volume averaged electrocardiograms were recorded from the transmural wall of ventricular wedge preparations ( n = 7) at baseline (4 mM potassium), hyperkalemia (8-12 mM), and hyperkalemia + Ca 2+ (3.6 mM). Isolated myocytes were studied during hyperkalemia (8 mM) and after Ca 2+ treatment (6 mM) to determine cellular RMP. MAIN RESULTS: Hyperkalemia markedly slowed conduction velocity (CV, by 67% ± 7%; p < 0.001) and homogeneously shortened action potential duration (APD, by 20% ± 10%; p < 0.002). In all preparations, this resulted in QRS widening and the "sine wave" pattern observed in severe hyperkalemia. Ca 2+ treatment restored CV (increase by 44% ± 18%; p < 0.02), resulting in narrowing of the QRS and normalization of the electrocardiogram, but did not restore APD. RMP was significantly elevated by hyperkalemia; however, it was not restored with Ca 2+ treatment suggesting a mechanism unrelated to "membrane stabilization." In addition, the effect of Ca 2+ was attenuated during L-type Ca 2+ channel blockade, suggesting a mechanism related to Ca 2+ -dependent (rather than normally sodium-dependent) conduction. CONCLUSIONS: These data suggest that Ca 2+ treatment for hyperkalemia restores conduction through Ca 2+ -dependent propagation, rather than restoration of membrane potential or "membrane stabilization." Our findings provide a mechanistic rationale for Ca 2+ treatment when hyperkalemia produces abnormalities of conduction (i.e., QRS prolongation).


Asunto(s)
Calcio , Hiperpotasemia , Hiperpotasemia/tratamiento farmacológico , Animales , Perros , Calcio/metabolismo , Potenciales de Acción/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Electrocardiografía , Membrana Celular/efectos de los fármacos
4.
J Bioenerg Biomembr ; 56(5): 483-493, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39266925

RESUMEN

Neurons of the subpostremal nucleus of the solitary tract (NTS) respond to changes in extracellular glucose with alterations in membrane potential with both depolarization and hyperpolarization. From 5 mM glucose, a rapid shift to 0.5 mM glucose produces a membrane depolarization by an unknown mechanism in most neurons. However, the mechanism involved in this response needs to be known. Here, we investigated if the low glucose-induced depolarization could be mimicked by reducing ATP synthesis and possible mediators of this effect. We showed that applying the mitochondrial uncoupler CCCP (1 µM) reproduced the effects of low glucose depolarizing the membrane, generating an inward current, and decreasing membrane resistance. On the other hand, activation of AMPK did not alter these parameters. To test if low glucose and CCCP could depolarize the membrane by affecting the ionic gradient, we inhibited the electrogenic Na/K pump with 10 µM of ouabain. We observed a similar membrane depolarization but not a decrease in membrane resistance. We conclude that perfusion of neurons of the subpostremal NTS with a low glucose solution depolarizes the membrane by probably reducing intracellular ATP, but not by activating AMPK or decreasing the ionic gradient across the membrane.


Asunto(s)
Adenosina Trifosfato , Glucosa , Mitocondrias , Neuronas , Núcleo Solitario , Animales , Ratas , Glucosa/metabolismo , Glucosa/farmacología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/biosíntesis , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Núcleo Solitario/metabolismo , Núcleo Solitario/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos
5.
Commun Biol ; 7(1): 1181, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300259

RESUMEN

A major driver of neuronal hyperexcitability is dysfunction of K+ channels, including voltage-gated KCNQ2/3 channels. Their hyperpolarized midpoint of activation and slow activation and deactivation kinetics produce a current that regulates membrane potential and impedes repetitive firing. Inherited mutations in KCNQ2 and KCNQ3 are linked to a wide spectrum of neurodevelopmental disorders (NDDs), ranging from benign familial neonatal seizures to severe epileptic encephalopathies and autism spectrum disorders. However, the impact of these variants on the molecular mechanisms underlying KCNQ3 channel function remains poorly understood and existing treatments have significant side effects. Here, we use voltage clamp fluorometry, molecular dynamic simulations, and electrophysiology to investigate NDD-associated variants in KCNQ3 channels. We identified two distinctive mechanisms by which loss- and gain-of function NDD-associated mutations in KCNQ3 affect channel gating: one directly affects S4 movement while the other changes S4-to-pore coupling. MD simulations and electrophysiology revealed that polyunsaturated fatty acids (PUFAs) primarily target the voltage-sensing domain in its activated conformation and form a weaker interaction with the channel's pore. Consistently, two such compounds yielded partial and complete functional restoration in R227Q- and R236C-containing channels, respectively. Our results reveal the potential of PUFAs to be developed into therapies for diverse KCNQ3-based channelopathies.


Asunto(s)
Canal de Potasio KCNQ3 , Simulación de Dinámica Molecular , Mutación , Trastornos del Neurodesarrollo , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/metabolismo , Humanos , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/tratamiento farmacológico , Activación del Canal Iónico/efectos de los fármacos , Animales , Células HEK293 , Potenciales de la Membrana/efectos de los fármacos
6.
Biochim Biophys Acta Biomembr ; 1866(8): 184386, 2024 12.
Artículo en Inglés | MEDLINE | ID: mdl-39343086

RESUMEN

We show that voltage alone can inactivate alamethicin channels, which has been previously observed for monazomycin and suzukacillin channels. The voltage required to trigger inactivation is above the potential to form channels, and, like with channel activation, this threshold reduces with increasing peptide concentration and membrane fluidity. Since similar monazomycin channels inactivate via channel break up and translocation, we hypothesized that inactivation of alamethicin channels occurs via the same mechanism. Our data prove this hypothesis to be true through two experiments. First, we show that inactivation of channels at positive voltages when peptides are supplied to only the cis side correlates to new channel activity on the trans side at negative potentials. This result indicates translocation of alamethicin peptides occurs only during voltage-induced inactivation. Second, we measured the ratio of steady-state (with inactivation) to ideal (without inactivation) conductance versus voltage for membranes with equal amounts of alamethicin on both sides and used these values to quantify alamethicin flux. Plotting flux versus steady-state conductance across multiple alamethicin concentrations shows a single linear dependence, signifying that translocated peptides originate from active channels that break up under prolonged voltage. Given the frequent use of alamethicin as model ion channels, these results add important understanding of their kinetic responses when subjected to prolonged, high voltages.


Asunto(s)
Alameticina , Alameticina/farmacología , Alameticina/metabolismo , Canales Iónicos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos
7.
Microbiol Res ; 289: 127918, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39342747

RESUMEN

Ketoconazole is a classical antifungal drug commonly used in the clinic. With the increased use of ketoconazole in recent years, an increasing number of drug-resistant strains have emerged during clinical treatment. It is well known that fungi acquire drug resistance in multiple ways, while the molecular mechanisms underlying ketoconazole resistance remain for comprehensive exploration. In this study, we found that the expression of the small plasma membrane protein-encoding gene PMP3 was significantly down-regulated in several clinically isolated ketoconazole-resistant strains, indicating the relationship between PMP3 expression and ketoconazole resistance. By knocking out the PMP3, we found that the absence of the Pmp3 resulted in a significant increase in resistance of Candida albicans to ketoconazole, which was also confirmed in a systemic infection model in mice. We further demonstrated that various physiological properties, such as cell membrane fluidity, plasma membrane potential, permeability and ergosterol distribution were altered in the pmp3Δ/Δ mutant, which is associated with the enhanced cellular resistance to ketoconazole. In addition, overexpression rather than deletion of PMP3 alters the hyphal development and biofilm formation capacity in C. albicans. This study reveals the contribution of Pmp3 to alteration of drug resistance in fungal pathogens, which may guide the development of novel antifungal strategies.


Asunto(s)
Antifúngicos , Candida albicans , Membrana Celular , Farmacorresistencia Fúngica , Proteínas Fúngicas , Cetoconazol , Pruebas de Sensibilidad Microbiana , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candida albicans/metabolismo , Cetoconazol/farmacología , Antifúngicos/farmacología , Animales , Farmacorresistencia Fúngica/genética , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Ratones , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Candidiasis/microbiología , Potenciales de la Membrana/efectos de los fármacos , Eliminación de Gen , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Hifa/crecimiento & desarrollo , Hifa/efectos de los fármacos , Hifa/genética , Ergosterol/metabolismo , Humanos , Modelos Animales de Enfermedad
8.
Am J Physiol Cell Physiol ; 327(4): C1023-C1034, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39159388

RESUMEN

Melatonin is synthesized in and secreted from the pineal glands and regulates circadian rhythms. Although melatonin has been reported to modulate the activity of ion channels in several tissues, its effects on pineal ion channels remain unclear. In the present study, the effects of melatonin on voltage-gated K+ (KV) channels, which play a role in regulating the resting membrane potential, were examined in rat pinealocytes. The application of melatonin reduced pineal KV currents in a concentration-dependent manner (IC50 = 309 µM). An expression analysis revealed that KV4.2 channels were highly expressed in rat pineal glands. Melatonin-sensitive currents were abolished by the small interfering RNA knockdown of KV4.2 channels in rat pinealocytes. In human embryonic kidney 293 (HEK293) cells expressing KV4.2 channels, melatonin decreased outward currents (IC50 = 479 µM). Inhibitory effects were mediated by a shift in the voltage dependence of steady-state inactivation in a hyperpolarizing direction. This inhibition was observed even in the presence of 100 nM luzindole, an antagonist of melatonin receptors. Melatonin also blocked the activity of KV4.3, KV1.1, and KV1.5 channels in reconstituted HEK293 cells. The application of 1 mM melatonin caused membrane depolarization in rat pinealocytes. Furthermore, KV4.2 channel inhibition by 5 mM 4-aminopyridine attenuated melatonin secretion induced by 1 µM noradrenaline in rat pineal glands. These results strongly suggest that melatonin directly inhibited KV4.2 channels and caused membrane depolarization in pinealocytes, resulting in a decrease in melatonin secretion through parasympathetic signaling pathway. This mechanism may function as a negative-feedback mechanism of melatonin secretion in pineal glands. NEW & NOTEWORTHY Melatonin is a hormone that is synthesized in and secreted from the pineal glands, which regulates circadian rhythms. However, the effects of melatonin on pineal ion channels remain unclear. The present study demonstrated that melatonin directly inhibited voltage-gated potassium KV4.2 channels, which are highly expressed in rat pinealocytes, and induced membrane depolarization, resulting in a decrease in melatonin secretion. This mechanism may function as a negative-feedback mechanism of melatonin secretion in pineal glands.


Asunto(s)
Melatonina , Glándula Pineal , Canales de Potasio Shal , Animales , Glándula Pineal/metabolismo , Glándula Pineal/efectos de los fármacos , Melatonina/farmacología , Humanos , Células HEK293 , Ratas , Masculino , Canales de Potasio Shal/metabolismo , Canales de Potasio Shal/genética , Ratas Sprague-Dawley , Potenciales de la Membrana/efectos de los fármacos
9.
Artículo en Inglés | MEDLINE | ID: mdl-39111513

RESUMEN

TMEM16A, a member of the Transmembrane protein 16 family, serves as the molecular basis for calcium activated chloride channels (CaCCs). We use RT-PCR to demonstrate the expression of TMEM16A in the neurons of Helicoverpa armigera, and record the CaCCs current of acute isolated neurons of H. armigera for the first time using patch clamp technology. In order to screen effective inhibitors of calcium-activated chloride channels, the inhibitory effects of four chloride channel inhibitors, CaCCinh-A01, NPPB, DIDS, and SITS, on CaCCs were compared. The inhibitory effects of the four inhibitors on the outward current of CaCCs were CaCCinh-A01 (10 µM, 56.31 %), NPPB (200 µM, 43.69 %), SITS (1 mM, 12.41 %) and DIDS (1 mM, 13.29 %). Among these inhibitors, CaCCinh-A01 demonstrated the highest efficacy as a blocker. To further explore whether calcium channel proteins can serve as potential targets of pyrethroids, we compared the effects of (type I) tefluthrin and (type II) deltamethrin on CaCCs. 10 µM and 100 µM tefluthrin can stimulate a large tail current in CaCCs, prolonging their deactivation time by 10.44 ms and 31.49 ms, and the V0.5 shifted in the hyperpolarization by 2-8 mV. Then, deltamethrin had no obvious effect on the deactivation and activation of CaCCs. Therefore, CaCCs of H. armigera can be used as a potential target of pyrethroids, but type I and type II pyrethroids have different effects on CaCCs.


Asunto(s)
Canales de Cloruro , Insecticidas , Mariposas Nocturnas , Neuronas , Piretrinas , Animales , Insecticidas/toxicidad , Insecticidas/farmacología , Piretrinas/toxicidad , Piretrinas/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Canales de Cloruro/metabolismo , Canales de Cloruro/antagonistas & inhibidores , Mariposas Nocturnas/efectos de los fármacos , Anoctamina-1/metabolismo , Anoctamina-1/antagonistas & inhibidores , Proteínas de Insectos/metabolismo , Proteínas de Insectos/antagonistas & inhibidores , Proteínas de Insectos/genética , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Nitrobenzoatos/farmacología , Helicoverpa armigera , Ciclopropanos , Hidrocarburos Fluorados
10.
Artículo en Inglés | MEDLINE | ID: mdl-39154976

RESUMEN

The gram-negative toxin lipopolysaccharides (LPS) are known to trigger inflammatory cytokines in mammals, which can result in pathological responses. Upon treatment of bacterial sepsis with antibiotics, the lysing bacteria can present a surge in LPS, inducing a cytokine storm. However, LPS can also have direct cellular effects, including transient rapid hyperpolarizing of the membrane potential, blocking glutamate receptors and even promoting release of glutamate. The detailed mechanism of action for these immediate responses is still unresolved. In addressing the membrane hyperpolarization, voltage gated K+ channel blockers 4-aminopyridine (4-AP, 3 mM), quinidine hydrochloride monohydrate (0.1 mM) and tetraethylammonium (TEA, 20 mM) were examined along with RNAi knockdowns of potential calcium activated K+ channels. The immediate responses of LPS were not blocked. Even in the presence of glutamate, the membrane still hyperpolarizes with LPS. When the driving gradient for the ionotropic glutamate receptors is enhanced during hyperpolarization, spontaneous quantal responses are dampened in amplitude. Thus, glutamate receptors are blocked, and the mechanism of hyperpolarization remains unresolved. The larval Drosophila glutamatergic neuromuscular junction is used as a model synaptic preparation to address the direct rapid actions by LPS.


Asunto(s)
Lipopolisacáridos , Potenciales de la Membrana , Animales , Lipopolisacáridos/farmacología , Potenciales de la Membrana/efectos de los fármacos , Canales de Potasio Calcio-Activados/metabolismo , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Drosophila melanogaster , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Larva/efectos de los fármacos , Larva/metabolismo
11.
Int J Mol Sci ; 25(16)2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39201487

RESUMEN

Previous studies have suggested a role for selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine (Prozac®) in the treatment of dizziness and inner ear vestibular dysfunction. The potential mechanism of action within the vestibular system remains unclear; however, fluoxetine has been reported to block certain types of K+ channel in other systems. Here, we investigated the direct actions of fluoxetine on membrane currents in presynaptic hair cells and postsynaptic calyx afferents of the gerbil peripheral vestibular system using whole cell patch clamp recordings in crista slices. We explored differences in K+ currents in peripheral zone (PZ) and central zone (CZ) calyces of the crista and their response to fluoxetine application. Outward K+ currents in PZ calyces showed greater inactivation at depolarized membrane potentials compared to CZ calyces. The application of 100 µM fluoxetine notably reduced K+ currents in calyx terminals within both zones of the crista, and the remaining currents exhibited distinct traits. In PZ cells, fluoxetine inhibited a non-inactivating K+ current and revealed a rapidly activating and inactivating K+ current, which was sensitive to blocking by 4-aminopyridine. This was in contrast to CZ calyces, where low-voltage-activated and non-inactivating K+ currents persisted following application of 100 µM fluoxetine. Additionally, marked inhibition of transient inward Na+ currents by fluoxetine was observed in calyces from both crista zones. Different concentrations of fluoxetine were tested, and the EC50 values were found to be 40 µM and 32 µM for K+ and Na+ currents, respectively. In contrast, 100 µM fluoxetine had no impact on voltage-dependent K+ currents in mechanosensory type I and type II vestibular hair cells. In summary, micromolar concentrations of fluoxetine are expected to strongly reduce both Na+ and K+ conductance in afferent neurons of the peripheral vestibular system in vivo. This would lead to inhibition of action potential firing in vestibular sensory neurons and has therapeutic implications for disorders of balance.


Asunto(s)
Fluoxetina , Gerbillinae , Fluoxetina/farmacología , Animales , Potenciales de la Membrana/efectos de los fármacos , Vestíbulo del Laberinto/efectos de los fármacos , Vestíbulo del Laberinto/metabolismo , Técnicas de Placa-Clamp , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Canales de Potasio/metabolismo , Masculino , Células Ciliadas Vestibulares/efectos de los fármacos , Células Ciliadas Vestibulares/metabolismo
12.
Nat Commun ; 15(1): 6877, 2024 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-39128925

RESUMEN

The bactericidal activity of several antibiotics partially relies on the production of reactive oxygen species (ROS), which is generally linked to enhanced respiration and requires the Fenton reaction. Bacterial persister cells, an important cause of recurring infections, are tolerant to these antibiotics because they are in a dormant state. Here, we use Bacillus subtilis cells in stationary phase, as a model system of dormant cells, to show that pharmacological induction of membrane depolarization enhances the antibiotics' bactericidal activity and also leads to ROS production. However, in contrast to previous studies, this results primarily in production of superoxide radicals and does not require the Fenton reaction. Genetic analyzes indicate that Rieske factor QcrA, the iron-sulfur subunit of respiratory complex III, seems to be a primary source of superoxide radicals. Interestingly, the membrane distribution of QcrA changes upon membrane depolarization, suggesting a dissociation of complex III. Thus, our data reveal an alternative mechanism by which antibiotics can cause lethal ROS levels, and may partially explain why membrane-targeting antibiotics are effective in eliminating persisters.


Asunto(s)
Antibacterianos , Bacillus subtilis , Membrana Celular , Especies Reactivas de Oxígeno , Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/metabolismo , Bacillus subtilis/fisiología , Especies Reactivas de Oxígeno/metabolismo , Antibacterianos/farmacología , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Superóxidos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Complejo III de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética
13.
Toxins (Basel) ; 16(8)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39195769

RESUMEN

Crotalphine is an analgesic peptide identified from the venom of the South American rattlesnake Crotalus durissus terrificus. Although its antinociceptive effect is well documented, its direct mechanisms of action are still unclear. The aim of the present work was to study the action of the crotalid peptide on the NaV1.7 channel subtype, a genetically validated pain target. To this purpose, the effects of crotalphine were evaluated on the NaV1.7 component of the tetrodotoxin-sensitive Na+ current in the dorsal root ganglion neurons of adult mice, using the whole-cell patch-clamp configuration, and on cell viability, using propidium iodide fluorescence and trypan blue assays. The results show that 18.7 µM of peptide inhibited 50% of the Na+ current. The blocking effect occurred without any marked change in the current activation and inactivation kinetics, but it was more important as the membrane potential was more positive. In addition, crotalphine induced an increase in the leakage current amplitude of approximately 150% and led to a maximal 31% decrease in cell viability at a high 50 µM concentration. Taken together, these results point out, for the first time, the effectiveness of crotalphine in acting on the NaV1.7 channel subtype, which may be an additional target contributing to the peptide analgesic properties and, also, although less efficiently, on a second cell plasma membrane component, leading to cell loss.


Asunto(s)
Analgésicos , Ganglios Espinales , Canal de Sodio Activado por Voltaje NAV1.7 , Neuronas , Tetrodotoxina , Animales , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/citología , Neuronas/efectos de los fármacos , Ratones , Tetrodotoxina/farmacología , Analgésicos/farmacología , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Venenos de Crotálidos/toxicidad , Venenos de Crotálidos/farmacología , Masculino , Crotalus , Potenciales de la Membrana/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Péptidos
14.
Cell Mol Life Sci ; 81(1): 374, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39210039

RESUMEN

Lysophosphatidylcholine (LPC) is a bioactive lipid present at high concentrations in inflamed and injured tissues where it contributes to the initiation and maintenance of pain. One of its important molecular effectors is the transient receptor potential canonical 5 (TRPC5), but the explicit mechanism of the activation is unknown. Using electrophysiology, mutagenesis and molecular dynamics simulations, we show that LPC-induced activation of TRPC5 is modulated by xanthine ligands and depolarizing voltage, and involves conserved residues within the lateral fenestration of the pore domain. Replacement of W577 with alanine (W577A) rendered the channel insensitive to strong depolarizing voltage, but LPC still activated this mutant at highly depolarizing potentials. Substitution of G606 located directly opposite position 577 with tryptophan rescued the sensitivity of W577A to depolarization. Molecular simulations showed that depolarization widens the lower gate of the channel and this conformational change is prevented by the W577A mutation or removal of resident lipids. We propose a gating scheme in which depolarizing voltage and lipid-pore helix interactions act together to promote TRPC5 channel opening.


Asunto(s)
Lisofosfatidilcolinas , Simulación de Dinámica Molecular , Canales Catiónicos TRPC , Humanos , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/química , Lisofosfatidilcolinas/metabolismo , Lisofosfatidilcolinas/farmacología , Animales , Activación del Canal Iónico/efectos de los fármacos , Células HEK293 , Mutación , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Potenciales de la Membrana/efectos de los fármacos
15.
FEMS Microbiol Lett ; 3712024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-39138064

RESUMEN

Garviecin LG34 produced by Lactococcus garvieae LG34 exhibits wide-spectrum antibacterial activity against both Gram-positive and Gram-negative bacteria. This work aimed at clarifying the antibacterial mode of action of garviecin LG34 against Gram-negative bacterium Salmonella typhimurium. To determine the concentration for the bacteriocin antimicrobial mode experiments, the minimum inhibitory concentration of garviecin LG34 against S. typhimurium CICC21484 was determined as 0.25 mg/ml. Garviecin LG34 decreased the viable count of S. typhimurium CICC21484 and its antibacterial activity was the dose and time dependant. Garviecin LG34 led to the dissipation of transmembrane potential, the rise in the extracellular conductivity, UV-absorbing material at 260 nm, and LDH level of S. typhimurium CICC21484. Scanning electron micrographs results shown that garviecin LG34 cause dramatic deformation and fragmentation including the flagellum shedding, pores formation in surface, and even completely breakage of S. typhimurium cell. Moreover, garviecin LG34 decreased the intracellular ATP level. The results of this study demonstrated that garviecin LG34 can destroy cell structure, increase membrane permeability of S. typhimurium, thereby might be used as biopreservative for treating food borne and salmonellosis resulting from Gram-negative bacterium S. typhimurium.


Asunto(s)
Antibacterianos , Pruebas de Sensibilidad Microbiana , Salmonella typhimurium , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/crecimiento & desarrollo , Antibacterianos/farmacología , Antibacterianos/química , Bacteriocinas/farmacología , Lactococcus/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Potenciales de la Membrana/efectos de los fármacos
16.
Cell Chem Biol ; 31(7): 1305-1323.e9, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39029456

RESUMEN

K2P potassium channels regulate excitability by affecting cellular resting membrane potential in the brain, cardiovascular system, immune cells, and sensory organs. Despite their important roles in anesthesia, arrhythmia, pain, hypertension, sleep, and migraine, the ability to control K2P function remains limited. Here, we describe a chemogenetic strategy termed CATKLAMP (covalent activation of TREK family K+ channels to clamp membrane potential) that leverages the discovery of a K2P modulator pocket site that reacts with electrophile-bearing derivatives of a TREK subfamily small-molecule activator, ML335, to activate the channel irreversibly. We show that CATKLAMP can be used to probe fundamental aspects of K2P function, as a switch to silence neuronal firing, and is applicable to all TREK subfamily members. Together, our findings exemplify a means to alter K2P channel activity that should facilitate molecular and systems level studies of K2P function and enable the search for new K2P modulators.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem , Humanos , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Animales , Células HEK293 , Ratones , Potenciales de la Membrana/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratas
17.
Crit Care Med ; 52(10): 1499-1508, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39046789

RESUMEN

OBJECTIVES: Hyperkalemia is a common life-threatening condition causing severe electrophysiologic derangements and arrhythmias. The beneficial effects of calcium (Ca 2+ ) treatment for hyperkalemia have been attributed to "membrane stabilization," by restoration of resting membrane potential (RMP). However, the underlying mechanisms remain poorly understood. Our objective was to investigate the mechanisms underlying adverse electrophysiologic effects of hyperkalemia and the therapeutic effects of Ca 2+ treatment. DESIGN: Controlled experimental trial. SETTING: Laboratory investigation. SUBJECTS: Canine myocytes and tissue preparations. INTERVENTIONS AND MEASUREMENTS: Optical action potentials and volume averaged electrocardiograms were recorded from the transmural wall of ventricular wedge preparations ( n = 7) at baseline (4 mM potassium), hyperkalemia (8-12 mM), and hyperkalemia + Ca 2+ (3.6 mM). Isolated myocytes were studied during hyperkalemia (8 mM) and after Ca 2+ treatment (6 mM) to determine cellular RMP. MAIN RESULTS: Hyperkalemia markedly slowed conduction velocity (CV, by 67% ± 7%; p < 0.001) and homogeneously shortened action potential duration (APD, by 20% ± 10%; p < 0.002). In all preparations, this resulted in QRS widening and the "sine wave" pattern observed in severe hyperkalemia. Ca 2+ treatment restored CV (increase by 44% ± 18%; p < 0.02), resulting in narrowing of the QRS and normalization of the electrocardiogram, but did not restore APD. RMP was significantly elevated by hyperkalemia; however, it was not restored with Ca 2+ treatment suggesting a mechanism unrelated to "membrane stabilization." In addition, the effect of Ca 2+ was attenuated during L-type Ca 2+ channel blockade, suggesting a mechanism related to Ca 2+ -dependent (rather than normally sodium-dependent) conduction. CONCLUSIONS: These data suggest that Ca 2+ treatment for hyperkalemia restores conduction through Ca 2+ -dependent propagation, rather than restoration of membrane potential or "membrane stabilization." Our findings provide a mechanistic rationale for Ca 2+ treatment when hyperkalemia produces abnormalities of conduction (i.e., QRS prolongation).


Asunto(s)
Calcio , Hiperpotasemia , Hiperpotasemia/tratamiento farmacológico , Animales , Perros , Calcio/metabolismo , Potenciales de Acción/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Electrocardiografía , Membrana Celular/efectos de los fármacos
18.
Biochem Pharmacol ; 227: 116421, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38996933

RESUMEN

Muscarinic receptors are G protein-coupled receptors (GPCRs) that play a role in various physiological functions. Previous studies have shown that these receptors, along with other GPCRs, are voltage-sensitive; both their affinity toward agonists and their activation are regulated by membrane potential. To our knowledge, whether the effect of antagonists on these receptors is voltage-dependent has not yet been studied. In this study, we used Xenopus oocytes expressing the M2 muscarinic receptor (M2R) to investigate this question. Our results indicate that the potencies of two M2R antagonists, atropine and scopolamine, are voltage-dependent; they are more effective at resting potential than under depolarization. In contrast, the M2R antagonist AF-DX 386 did not exhibit voltage-dependent potency.Furthermore, we discovered that the voltage dependence of M2R activation by acetylcholine remains unchanged in the presence of two allosteric modulators, the negative modulator gallamine and the positive modulator LY2119620. These findings enhance our understanding of GPCRs' voltage dependence and may have pharmacological implications.


Asunto(s)
Antagonistas Muscarínicos , Oocitos , Receptor Muscarínico M2 , Xenopus laevis , Animales , Receptor Muscarínico M2/antagonistas & inhibidores , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M2/agonistas , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Antagonistas Muscarínicos/farmacología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oocitos/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Atropina/farmacología , Escopolamina/farmacología , Acetilcolina/metabolismo , Acetilcolina/farmacología , Femenino , Sulfonamidas , Tiadiazoles
19.
Inflamm Res ; 73(9): 1459-1476, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38965133

RESUMEN

OBJECTIVE: We aimed to broaden our understanding of a potential interaction between B1R and TLR4, considering earlier studies suggesting that lipopolysaccharide (LPS) may trigger B1R stimulation. METHODS: We assessed the impact of DBK and LPS on the membrane potential of thoracic aortas from C57BL/6, B1R, or TLR4 knockout mice. Additionally, we examined the staining patterns of these receptors in the thoracic aortas of C57BL/6 and in endothelial cells (HBMEC). RESULTS: DBK does not affect the resting membrane potential of aortic rings in C57BL/6 mice, but it hyperpolarizes preparations in B1KO and TLR4KO mice. The hyperpolarization mechanism in B1KO mice involves B2R, and the TLR4KO response is independent of cytoplasmic calcium influx but relies on potassium channels. Conversely, LPS hyperpolarizes thoracic aorta rings in both C57BL/6 and B1KO mice, with the response unaffected by a B1R antagonist. Interestingly, the absence of B1R alters the LPS response to potassium channels. These activities are independent of nitric oxide synthase (NOS). While exposure to DBK and LPS does not alter B1R and TLR4 mRNA expression, treatment with these agonists increases B1R staining in endothelial cells of thoracic aortic rings and modifies the staining pattern of B1R and TLR4 in endothelial cells. Proximity ligation assay suggests a interaction between the receptors. CONCLUSION: Our findings provide additional support for a putative connection between B1R and TLR4 signaling. Given the involvement of these receptors and their agonists in inflammation, it suggests that drugs and therapies targeting their effects could be promising therapeutic avenues worth exploring.


Asunto(s)
Aorta Torácica , Células Endoteliales , Lipopolisacáridos , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Bradiquinina B1 , Receptor Toll-Like 4 , Animales , Masculino , Ratones , Aorta Torácica/metabolismo , Bradiquinina/farmacología , Bradiquinina/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Potenciales de la Membrana/efectos de los fármacos , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B1/genética , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genética , Femenino
20.
Foodborne Pathog Dis ; 21(7): 447-457, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38985570

RESUMEN

Bacillus cereus causes food poisoning by producing toxins that cause diarrhea and vomiting and, in severe cases, endocarditis, meningitis, and other diseases. It also tends to form biofilms and spores that lead to contamination of the food production environment. Citral is a potent natural antibacterial agent, but its antibacterial activity against B. cereus has not been extensively studied. In this study, we first determined the minimum inhibitory concentrations and minimum bactericidal concentrations, growth curves, killing effect in different media, membrane potential, intracellular adenosine triphosphate (ATP), reactive oxygen species levels, and morphology of vegetative cells, followed by germination rate, morphology, germination state of spores, and finally biofilm clearance effect. The results showed that the minimum inhibitory concentrations and minimum bactericidal concentrations of citral against bacteria ranged from 100 to 800 µg/mL. The lag phase of bacteria was effectively prolonged by citral, and the growth rate of bacteria was slowed down. Bacteria in Luria-Bertani broth were reduced to below the detection limit by citral at 800 µg/mL within 0.5 h. Bacteria in rice were reduced to 3 log CFU/g by citral at 4000 µg/mL within 0.5 h. After treatment with citral, intracellular ATP concentration was reduced, membrane potential was altered, intracellular reactive oxygen species concentration was increased, and normal cell morphology was altered. After treatment with citral at 400 µg/mL, spore germination rate was reduced to 16.71%, spore morphology was affected, and spore germination state was altered. It also had a good effect on biofilm removal. The present study showed that citral had good bacteriostatic activity against B. cereus vegetative cells and its spores and also had a good clearance effect on its biofilm. Citral has the potential to be used as a bacteriostatic substance for the control of B. cereus in food industry production.


Asunto(s)
Monoterpenos Acíclicos , Bacillus cereus , Biopelículas , Monoterpenos Acíclicos/farmacología , Antiinfecciosos/farmacología , Bacillus cereus/efectos de los fármacos , Bacillus cereus/crecimiento & desarrollo , Bacillus cereus/ultraestructura , Esporas Bacterianas/efectos de los fármacos , Biopelículas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Oryza/microbiología , Potenciales de la Membrana/efectos de los fármacos , Espacio Intracelular/enzimología , Adenosina Trifosfato/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Microscopía Electrónica de Rastreo , Microbiología de Alimentos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...