Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 226
Filtrar
1.
Exp Neurol ; 379: 114849, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38857748

RESUMO

Cerebral ischemic stroke is a serious disease with high mortality and disability rates. However, few neuroprotective drugs have been used for ischemic stroke in the clinic. Two main reasons may be responsible for this failure: difficulty in penetrating the blood-brain barrier (BBB) and easily inactivated in the blood circulation. Ferroptosis, a lipid oxidation-related cell death, plays significant roles in cerebral ischemia-reperfusion injury. We utilized RVG29, a peptide derived from Rabies virus glycoprotein, to obtain BBB-targeted lipid nanoparticles (T-LNPs) in order to investigate whether T-LNPs improved the neuroprotective effects of Ferrostatin-1 (Fer1, an inhibitor of ferroptosis) against cerebral ischemic damage. T-LNPs significantly increased BBB penetration following oxygen/glucose deprivation exposure in an in vitro BBB model and enhanced the fluorescence distribution in brain tissues at 6 h post-administration in a cerebral ischemic murine model. Moreover, T-LNPs encapsulated Fer1 (T-LNPs-Fer1) significantly enhanced the inhibitory effects of Fer1 on ferroptosis by maintaining the homeostasis of NADPH oxidase 4 (NOX4) and glutathione peroxidase 4 (GPX4) signals in neuronal cells after cerebral ischemia. T-LNPs-Fer1 significantly suppressed oxidative stress [heme oxygenase-1 expression and malondialdehyde (the product of lipid ROS reaction)] in neurons and alleviated ischemia-induced neuronal cell death, compared to Fer1 alone without encapsulation. Furthermore, T-LNPs-Fer1 significantly reduced cerebral infarction and improved behavior functions compared to Fer1-treated cerebral ischemic mice after 45-min ischemia/24-h reperfusion. These findings showed that the T-LNPs helped Fer1 penetrate the BBB and improved the neuroprotection of Fer1 against cerebral ischemic damage in experimental stroke, providing a feasible translational strategy for the development of clinical drugs for the treatment of ischemic stroke.


Assuntos
Barreira Hematoencefálica , Cicloexilaminas , Nanopartículas , Fármacos Neuroprotetores , Fenilenodiaminas , Animais , Camundongos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Fármacos Neuroprotetores/farmacologia , Nanopartículas/administração & dosagem , Masculino , Cicloexilaminas/farmacologia , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/patologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Isquemia Encefálica/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Ferroptose/efeitos dos fármacos , Glicoproteínas , Lipossomos , Fragmentos de Peptídeos , Proteínas Virais
2.
Eur J Pharmacol ; 976: 176699, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38825302

RESUMO

Clinically, statins have long been used for the prevention and treatment of chronic renal diseases, however, the underlying mechanisms are not fully elucidated. The present study investigated the effects of atorvastatin on diabetes renal injury and ferroptosis signaling. A mouse model of diabetes was established by the intraperitoneal injection of streptozotocin (50 mg/kg/day) plus a high fat diet with or without atorvastatin treatment. Diabetes mice manifested increased plasma glucose and lipid profile, proteinuria, renal injury and fibrosis, atorvastatin significantly lowered plasma lipid profile, proteinuria, renal injury in diabetes mice. Atorvastatin reduced renal reactive oxygen species (ROS), iron accumulation and renal expression of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), transferrin receptor 1 (TFR1), and increased renal expression of glutathione peroxidase 4 (GPX4), nuclear factor erythroid 2-related factor (NRF2) and ferritin heavy chain (FTH) in diabetes mice. Consistent with the findings in vivo, atorvastatin prevented high glucose-induced ROS formation and Fe2+ accumulation, an increase in the expression of 4-HNE, MDA and TFR1, and a decrease in cell viability and the expression of NRF2, GPX4 and FTH in HK2 cells. Atorvastatin also reversed ferroptosis inducer erastin-induced ROS production, intracellular Fe2+ accumulation and the changes in the expression of above-mentioned ferroptosis signaling molecules in HK2 cells. In addition, atorvastatin alleviated high glucose- or erastin-induced mitochondria injury. Ferroptosis inhibitor ferrostatin-1 and antioxidant N-acetylcysteine (NAC) equally reversed the expression of high glucose-induced ferroptosis signaling molecules. Our data support the notion that statins can inhibit diabetes-induced renal oxidative stress and ferroptosis, which may contribute to statins protection of diabetic nephropathy.


Assuntos
Atorvastatina , Nefropatias Diabéticas , Ferroptose , Estresse Oxidativo , Espécies Reativas de Oxigênio , Transdução de Sinais , Ferroptose/efeitos dos fármacos , Animais , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , Masculino , Transdução de Sinais/efeitos dos fármacos , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/complicações , Camundongos Endogâmicos C57BL , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Linhagem Celular , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico
3.
Kaohsiung J Med Sci ; 40(8): 710-721, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38837857

RESUMO

The proinflammatory properties of high-mobility group box protein 1 (HMGB1) in sepsis have been extensively studied. This study aimed to investigate the impact of HMGB1 on ferroptosis and its molecular mechanism in sepsis-induced acute lung injury (ALI). A septic mouse model was established using the cecal ligation and puncture method. Blocking HMGB1 resulted in improved survival rates, reduced lung injury, decreased levels of ferroptosis markers (reactive oxygen species, malondialdehyde, and Fe2+), and enhanced antioxidant enzyme activities (superoxide dismutase and catalase) in septic mice. In addition, knockdown of HMGB1 reduced cellular permeability, ferroptosis markers, and raised antioxidant enzyme levels in lipopolysaccharide (LPS)-stimulated MLE-12 cells. Silencing of HMGB1 led to elevations in the expressions of ferroptosis core-regulators in LPS-treated MLE-12 cells, such as solute carrier family 7 member 11 (SLC7A11), solute carrier family 3 member A2 (SLC3A2), and glutathione peroxidase 4. Furthermore, blocking HMGB1 did not alter ferroptosis, oxidative stress-related changes, and permeability in LPS-treated MLE-12 cells that were pretreated with ferrostatin-1 (a ferroptosis inhibitor). HMGB1 inhibition also led to elevated expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream targets, heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) in LPS-treated MLE-12 cells and lung tissues from septic mice. The Nrf2-specific inhibitor ML385 reversed the effects of HMGB1 silencing on ferroptosis and cell permeability in LPS-treated MLE-12 cells. Our findings indicated that the inhibition of HMGB1 restrains ferroptosis and oxidative stress, thereby alleviating sepsis-induced ALI through the activation of Nrf2 signaling.


Assuntos
Lesão Pulmonar Aguda , Ferroptose , Proteína HMGB1 , Lipopolissacarídeos , Fator 2 Relacionado a NF-E2 , Estresse Oxidativo , Sepse , Transdução de Sinais , Animais , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Proteína HMGB1/metabolismo , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/genética , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/tratamento farmacológico , Ferroptose/efeitos dos fármacos , Sepse/complicações , Sepse/metabolismo , Sepse/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Camundongos , Lipopolissacarídeos/toxicidade , Transdução de Sinais/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Cicloexilaminas/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/antagonistas & inibidores , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Sistema y+ de Transporte de Aminoácidos
4.
J Affect Disord ; 359: 364-372, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38772507

RESUMO

Depression, a complex disorder with significant treatment challenges, necessitates innovative therapeutic approaches to address its multifaceted nature and enhance treatment outcomes. The modulation of KCNQ potassium (K+) channels, pivotal regulators of neuronal excitability and neurotransmitter release, is a promising innovative therapeutic target in psychiatry. Widely expressed across various tissues, including the nervous and cardiovascular systems, KCNQ channels play a crucial role in modulating membrane potential and regulating neuronal activity. Recent preclinical evidence suggests that KCNQ channels, particularly KCNQ3, contribute to the regulation of neuronal excitability within the reward circuitry, offering a potential target for alleviating depressive symptoms, notably anhedonia. Studies using animal models demonstrate that interventions targeting KCNQ channels can restore dopaminergic firing balance and mitigate depressive symptoms. Human studies investigating the effects of KCNQ channel activators, such as ezogabine, have shown promising results in alleviating depressive symptoms and anhedonia. The aforementioned observations underscore the therapeutic potential of KCNQ channel modulation in depression management and highlight the need and justification for phase 2 and phase 3 dose-finding studies as well as studies prespecifying symptomatic targets in depression including anhedonia.


Assuntos
Antidepressivos , Carbamatos , Transtorno Depressivo Maior , Canais de Potássio KCNQ , Fenilenodiaminas , Animais , Humanos , Anedonia/efeitos dos fármacos , Anedonia/fisiologia , Antidepressivos/uso terapêutico , Antidepressivos/farmacologia , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Transtorno Depressivo Maior/tratamento farmacológico , Canais de Potássio KCNQ/agonistas , Canais de Potássio KCNQ/metabolismo , Canal de Potássio KCNQ3/genética , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico
5.
Acta Pharmacol Sin ; 45(8): 1673-1685, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38641746

RESUMO

Acute kidney injury (AKI) is defined as sudden loss of renal function characterized by increased serum creatinine levels and reduced urinary output with a duration of 7 days. Ferroptosis, an iron-dependent regulated necrotic pathway, has been implicated in the progression of AKI, while ferrostatin-1 (Fer-1), a selective inhibitor of ferroptosis, inhibited renal damage, oxidative stress and tubular cell death in AKI mouse models. However, the clinical translation of Fer-1 is limited due to its lack of efficacy and metabolic instability. In this study we designed and synthesized four Fer-1 analogs (Cpd-A1, Cpd-B1, Cpd-B2, Cpd-B3) with superior plasma stability, and evaluated their therapeutic potential in the treatment of AKI. Compared with Fer-1, all the four analogs displayed a higher distribution in mouse renal tissue in a pharmacokinetic assay and a more effective ferroptosis inhibition in erastin-treated mouse tubular epithelial cells (mTECs) with Cpd-A1 (N-methyl-substituted-tetrazole-Fer-1 analog) being the most efficacious one. In hypoxia/reoxygenation (H/R)- or LPS-treated mTECs, treatment with Cpd-A1 (0.25 µM) effectively attenuated cell damage, reduced inflammatory responses, and inhibited ferroptosis. In ischemia/reperfusion (I/R)- or cecal ligation and puncture (CLP)-induced AKI mouse models, pre-injection of Cpd-A1 (1.25, 2.5, 5 mg·kg-1·d-1, i.p.) dose-dependently improved kidney function, mitigated renal tubular injury, and abrogated inflammation. We conclude that Cpd-A1 may serve as a promising therapeutic agent for the treatment of AKI.


Assuntos
Injúria Renal Aguda , Ferroptose , Camundongos Endogâmicos C57BL , Fenilenodiaminas , Animais , Ferroptose/efeitos dos fármacos , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Camundongos , Masculino , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Cicloexilaminas/farmacologia , Cicloexilaminas/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo
6.
Biomater Sci ; 11(7): 2395-2404, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36786425

RESUMO

Nitric oxide (NO) plays various physiologically favorable roles in the body. However, excessive production of NO causes inflammation and leads to various chronic inflammatory diseases. A typical NO-related disease is rheumatoid arthritis (RA), and it is well known that NO is a critical molecule for inflammation in the pathophysiology of RA. Therefore, depletion of NO can be an attractive treatment option for RA. In this study, we proposed a new method to discover effective NO scavengers in the form of small molecules. o-Phenylenediamine (o-PD), the core structure of the NO scavenger, is a diamino-aromatic compound that irreversibly reacts with NO through nucleophilic substitution of amine. Inspired by the nucleophilicity, we attempted to find new scavenger candidates by searching for conditions that increase the nucleophilicity of the amine moieties. Candidates were classified into the basic form o-PD, monoamine aniline, o-PD substituted with a nitro group, carboxyl group, and three methyl groups. The NO-scavenging ability of these candidates was demonstrated using the DAF-2 assay. N-Methyl-o-PD (N-Me) in the methyl (-CH3) group had the highest reactivity with NO among the candidates, and the efficiency of NO scavengers was confirmed in vitro and in vivo. Depleted levels of NO and reduced levels of pro-inflammatory cytokines by N-Me demonstrated remarkable therapeutic efficacy against joint damage and delayed severity in a collagen-induced arthritis (CIA) model. Therefore, our findings suggest that N-Me is a new NO scavenger with great potential for RA treatment and further clinical drug development.


Assuntos
Artrite Reumatoide , Óxido Nítrico , Humanos , Artrite Reumatoide/tratamento farmacológico , Inflamação/tratamento farmacológico , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Citocinas
7.
Brain ; 146(4): 1554-1560, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-36718088

RESUMO

Recurrent episodes of weakness in periodic paralysis are caused by intermittent loss of muscle fibre excitability, as a consequence of sustained depolarization of the resting potential. Repolarization is favoured by increasing the fibre permeability to potassium. Based on this principle, we tested the efficacy of retigabine, a potassium channel opener, to suppress the loss of force induced by a low-K+ challenge in hypokalaemic periodic paralysis (HypoPP). Retigabine can prevent the episodic loss of force in HypoPP. Knock-in mutant mouse models of HypoPP (Cacna1s p.R528H and Scn4a p.R669H) were used to determine whether pre-treatment with retigabine prevented the loss of force, or post-treatment hastened recovery of force for a low-K+ challenge in an ex vivo contraction assay. Retigabine completely prevents the loss of force induced by a 2 mM K+ challenge (protection) in our mouse models of HypoPP, with 50% inhibitory concentrations of 0.8 ± 0.13 µM and 2.2 ± 0.42 µM for NaV1.4-R669H and CaV1.1-R528H, respectively. In comparison, the effective concentration for the KATP channel opener pinacidil was 10-fold higher. Application of retigabine also reversed the loss of force (rescue) for HypoPP muscle maintained in 2 mM K+. Our findings show that retigabine, a selective agonist of the KV7 family of potassium channels, is effective for the prevention of low-K+ induced attacks of weakness and to enhance recovery from an ongoing loss of force in mouse models of type 1 (Cacna1s) and type 2 (Scn4a) HypoPP. Substantial protection from the loss of force occurred in the low micromolar range, well within the therapeutic window for retigabine.


Assuntos
Paralisia Periódica Hipopotassêmica , Camundongos , Animais , Músculo Esquelético , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico
8.
J Med Chem ; 65(16): 11340-11364, 2022 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-35972998

RESUMO

Neuronal Kv7 channels represent important pharmacological targets for hyperexcitability disorders including epilepsy. Retigabine is the prototype Kv7 activator clinically approved for seizure treatment; however, severe side effects associated with long-term use have led to its market discontinuation. Building upon the recently described cryoEM structure of Kv7.2 complexed with retigabine and on previous structure-activity relationship studies, a small library of retigabine analogues has been designed, synthesized, and characterized for their Kv7 opening ability using both fluorescence- and electrophysiology-based assays. Among all tested compounds, 60 emerged as a potent and photochemically stable neuronal Kv7 channel activator. Compared to retigabine, compound 60 displayed a higher brain/plasma distribution ratio, a longer elimination half-life, and more potent and effective anticonvulsant effects in an acute seizure model in mice. Collectively, these data highlight compound 60 as a promising lead compound for the development of novel Kv7 activators for the treatment of hyperexcitability diseases.


Assuntos
Anticonvulsivantes , Canal de Potássio KCNQ3 , Animais , Anticonvulsivantes/química , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Carbamatos , Canal de Potássio KCNQ2 , Camundongos , Fenilenodiaminas/química , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico
9.
Pharmacol Res Perspect ; 10(4): e00983, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35881020

RESUMO

Altered motor neuron excitability in patients with amyotrophic lateral sclerosis (ALS) has been suggested to be an early pathophysiological mechanism associated with motor neuron death. Compounds that affect membrane excitability may therefore have disease-modifying effects. Through which mechanism(s), these compounds modulate membrane excitability is mostly provided by preclinical studies, yet remains challenging to verify in clinical studies. Here, we investigated how retigabine affects human myelinated motor axons by applying computational modeling to interpret the complex excitability changes in a recent trial involving 18 ALS patients. Compared to baseline, the post-dose excitability differences were modeled well by a hyperpolarizing shift of the half-activation potential of slow potassium (K+ )-channels (till 2 mV). These findings verify that retigabine targets slow K+ -channel gating and highlight the usefulness of computational models. Further developments of this approach may facilitate the identification of early target engagement and ultimately aid selecting responders leading to more personalized treatment strategies.


Assuntos
Esclerose Lateral Amiotrófica , Esclerose Lateral Amiotrófica/tratamento farmacológico , Axônios/fisiologia , Carbamatos , Humanos , Neurônios Motores , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico
10.
Exp Neurol ; 355: 114141, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35691372

RESUMO

Anti-seizure drug (ASD) targets are widely expressed in both excitatory and inhibitory neurons. It remains unknown if the action of an ASD upon inhibitory neurons could counteract its beneficial effects on excitatory neurons (or vice versa), thereby reducing the efficacy of the ASD. Here, we examine whether the efficacy of the ASD retigabine (RTG) is altered after removal of the Kv7 potassium channel subunit KCNQ2, one of its drug targets, from parvalbumin-expressing interneurons (PV-INs). Parvalbumin-Cre (PV-Cre) mice were crossed with Kcnq2-floxed (Kcnq2fl/fl) mice to conditionally delete Kcnq2 from PV-INs. In these conditional knockout mice (cKO, PV-Kcnq2fl/fl), RTG (10 mg/kg, i.p.) significantly delayed the onset of either picrotoxin (PTX, 10 mg/kg, i.p)- or kainic acid (KA, 30 mg/kg, i.p.)-induced convulsive seizures compared to vehicle, while RTG was not effective in wild-type littermates (WT). Immunostaining for KCNQ2 and KCNQ3 revealed that both subunits were enriched at axon initial segments (AISs) of hippocampal CA1 PV-INs, and their specific expression was selectively abolished in cKO mice. Accordingly, the M-currents recorded from CA1 PV-INs and their sensitivity to RTG were significantly reduced in cKO mice. While the ability of RTG to suppress CA1 excitatory neurons in hippocampal slices was unchanged in cKO mice, its suppressive effect on the spike activity of CA1 PV-INs was significantly reduced compared with WT mice. In addition, the RTG-induced suppression on intrinsic membrane excitability of PV-INs in WT mice was significantly reduced in cKO mice. These findings suggest that preventing RTG from suppressing PV-INs improves its anticonvulsant effect.


Assuntos
Parvalbuminas , Fenilenodiaminas , Animais , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Interneurônios/metabolismo , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Parvalbuminas/metabolismo , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico
11.
CNS Drugs ; 36(3): 207-216, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35258812

RESUMO

Major depressive disorder (MDD) is a leading cause of disability worldwide and less than one-third of patients with MDD achieve stable remission of symptoms, despite currently available treatments. Although MDD represents a serious health problem, a complete understanding of the neurobiological mechanisms underlying this condition continues to be elusive. Accumulating evidence from preclinical and animal studies provides support for the antidepressant potential of modulators of KCNQ voltage-gated potassium (K+) channels. KCNQ K+ channels, through regulation of neuronal excitability and activity, contribute to neurophysiological mechanisms underlying stress resilience, and represent potential targets of drug discovery for depression. The present article focuses on the pharmacology and efficacy of KCNQ2/3 K+ channel openers as novel therapeutic agents for depressive disorders from initial studies conducted on animal models showing depressive-like behaviors to recent work in humans that examines the potential for KCNQ2/3 channel modulators as novel antidepressants. Data from preclinical work suggest that KCNQ-type K+ channels are an active mediator of stress resilience and KCNQ2/3 K+ channel openers show antidepressant efficacy. Similarly, evidence from clinical trials conducted in patients with MDD using the KCNQ2/3 channel opener ezogabine (retigabine) showed significant improvements in depressive symptoms and anhedonia. Overall, KCNQ channel openers appear a promising target for the development of novel therapeutics for the treatment of psychiatric disorders and specifically for MDD.


Assuntos
Transtorno Depressivo Maior , Canais de Potássio KCNQ , Animais , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Transtorno Depressivo Maior/tratamento farmacológico , Humanos , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico
12.
Neurosci Lett ; 771: 136460, 2022 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-35051437

RESUMO

Transcranial magnetic stimulation (TMS) is a neurophysiological technique that enables noninvasive evaluation of neuronal excitability in the brain. In the past, a large number of antiepileptic drugs were shown to increase the motor threshold (MT) in clinical TMS studies, suggesting the inhibition of excessive neuronal excitability. To facilitate drug development, the confirmation of similar changes in neurophysiological biomarkers in both preclinical and clinical studies is crucial; however, until now, there have been no data showing the drug efficacies on neuronal excitabilities as measured using TMS in rodents. In this study, we found that the antiepileptic drugs, lamotrigine (10 mg/kg) and retigabine (5 mg/kg), significantly increased the MT in rats using TMS, which is similar to clinical study findings. In addition, we demonstrated that these drugs could inhibit maximal electroshock (MES)-induced seizures in rats when given at the same dose required to be effective in the TMS experiment. These findings suggest that the effects of antiepileptic drugs in our rat TMS system have a similar sensitivity to that of the antiepileptic effects in rats with MES-induced seizures. The measurement of MT in a TMS study may be a noninvasive translational approach for predicting antiepileptic efficacy in drug development.


Assuntos
Anticonvulsivantes/uso terapêutico , Carbamatos/uso terapêutico , Potencial Evocado Motor , Lamotrigina/uso terapêutico , Fenilenodiaminas/uso terapêutico , Convulsões/tratamento farmacológico , Animais , Eletrochoque/efeitos adversos , Masculino , Ratos , Ratos Sprague-Dawley , Convulsões/etiologia , Estimulação Magnética Transcraniana
13.
Clin Sci (Lond) ; 136(1): 45-60, 2022 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-34918039

RESUMO

Cold storage/rewarming is an inevitable process for kidney transplantation from deceased donors, which correlates closely with renal ischemia-reperfusion injury (IRI) and the occurrence of delayed graft function. Histone deacetylases (HDAC) are important epigenetic regulators, but their involvement in cold storage/rewarming injury in kidney transplantation is unclear. In the present study, we showed a dynamic change of HDAC3 in a mouse model of kidney cold storage followed by transplantation. We then demonstrated that the selective HDAC3 inhibitor RGFP966 could reduce acute tubular injury and cell death after prolonged cold storage with transplantation. RGFP966 also improved renal function, kidney repair and tubular integrity when the transplanted kidney became the sole life-supporting graft in the recipient mouse. In vitro, cold storage of proximal tubular cells followed by rewarming induced remarkable cell death, which was suppressed by RGFP966 or knockdown of HDAC3 with shRNA. Inhibition of HDAC3 decreased the mitochondrial pathway of apoptosis and preserved mitochondrial membrane potential. Collectively, HDAC3 plays a pathogenic role in cold storage/rewarming injury in kidney transplantation, and its inhibition may be a therapeutic option.


Assuntos
Acrilamidas/uso terapêutico , Histona Desacetilases/efeitos dos fármacos , Transplante de Rim , Fenilenodiaminas/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Aloenxertos , Animais , Apoptose , Temperatura Baixa , Técnicas de Silenciamento de Genes , Histona Desacetilases/genética , Túbulos Renais Proximais/patologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Preservação de Órgãos/efeitos adversos , RNA Interferente Pequeno
14.
Neuropharmacology ; 203: 108884, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34785163

RESUMO

Epilepsy, one of the most common and most disabling neurological disorders, is characterized by spontaneous recurrent seizures, often associated with structural brain alterations and cognitive and psychiatric comorbidities. In about 30% of patients, the seizures are resistant to current treatments; so more effective treatments are urgently needed. Among the ∼30 clinically approved antiseizure drugs, retigabine (ezogabine) is the only drug that acts as a positive allosteric modulator (or opener) of voltage-gated Kv7 potassium channels, which is particularly interesting for some genetic forms of epilepsy. Here we describe a novel dual-mode-of-action compound, GRT-X (N-[(3-fluorophenyl)-methyl]-1-(2-methoxyethyl)-4-methyl-2-oxo-(7-trifluoromethyl)-1H-quinoline-3-carboxylic acid amide) that activates both Kv7 potassium channels and the mitochondrial translocator protein 18 kDa (TSPO), leading to increased synthesis of brain neurosteroids. TSPO activators are known to exert anti-inflammatory, neuroprotective, anxiolytic, and antidepressive effects, which, together with an antiseizure effect (mediated by Kv7 channels), would be highly relevant for the treatment of epilepsy. This prompted us to compare the antiseizure efficacy of retigabine and GRT-X in six mouse and rat models of epileptic seizures, including the 6-Hz model of difficult-to-treat focal seizures. Furthermore, the tolerability of the two compounds was compared in mice and rats. Potency comparisons were based on both doses and peak plasma concentrations. Overall, GRT-X was more effective than retigabine in three of the six seizure models used here, the most important difference being the high efficacy in the 6-Hz (32 mA) seizure model in mice. Based on drug plasma levels, GRT-X was at least 30 times more potent than retigabine in the latter model. These data indicate that GRT-X is a highly interesting novel anti-seizure drug with a unique (first-in-class) dual-mode mechanism of action.


Assuntos
Anticonvulsivantes/uso terapêutico , Carbamatos/uso terapêutico , Fenilenodiaminas/uso terapêutico , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Receptores de GABA/metabolismo , Convulsões/tratamento farmacológico , Convulsões/metabolismo , Animais , Anticonvulsivantes/química , Anticonvulsivantes/farmacologia , Células CHO , Carbamatos/farmacologia , Cricetulus , Relação Dose-Resposta a Droga , Eletrochoque/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Fenilenodiaminas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Convulsões/etiologia , Resultado do Tratamento
15.
Oral Dis ; 28(3): 611-620, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33503275

RESUMO

BACKGROUND: Cyclin-dependent kinase 7 (CDK7) has been critically linked to human cancer. However, the roles of CDK7 in head and neck squamous cell carcinoma (HNSCC) remain incompletely known. Here, we sought to dissect the functions of CDK7 underlying HNSCC tumorigenesis and explore whether pharmacological inhibition of CDK7 could induce anti-cancer effects. METHODS: CDK7 expression was measured in a panel of HNSCC cell lines with p53 mutation and 20 pairs of HNSCC samples and adjacent non-tumor tissues. Genetic targeting and pharmacological inhibition of CDK7 were conducted to dissect the biological roles of CDK7 in p53-mutated HNSCC cells. An HNSCC xenograft model was developed to determine the therapeutic effects of THZ1 in vivo. Potential genes and pathways responsible for therapeutic effects of THZ1 were identified by genome-wide RNA-sequencing and bioinformatics interrogations. RESULTS: CDK7 expression was significantly elevated in cancerous cells and samples as compared with their adjacent non-tumor counterparts. Impaired cell proliferation, migration, and invasion as well increased apoptosis were observed in cells upon CDK7 knockdown or THZ1 exposure. THZ1 administration potently inhibited tumor overgrowth in vivo. Mechanistically, hundreds of genes enriched in cell proliferation, apoptosis, and cancer-related categories were identified to be potentially mediated the therapeutic effects of THZ1 in HNSCC. CONCLUSION: Our findings reveal that CDK7 might serve as a novel putative pro-oncogenic gene underlying HNSCC tumorigenesis and therapeutic targeting of CDK7 might be a promising strategy for p53-mutated HNSCC.


Assuntos
Neoplasias de Cabeça e Pescoço , Proteína Supressora de Tumor p53 , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Quinases Ciclina-Dependentes/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/genética , Humanos , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Fosfotransferases (Aceptor do Grupo Álcool) , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Quinase Ativadora de Quinase Dependente de Ciclina
16.
J Am Soc Nephrol ; 32(11): 2759-2776, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34716241

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD), the most common inherited kidney disease, is regulated by different forms of cell death, including apoptosis and autophagy. However, the role in ADPKD of ferroptosis, a recently discovered form of cell death mediated by iron and lipid metabolism, remains elusive. METHODS: To determine a pathophysiologic role of ferroptosis in ADPKD, we investigated whether the absence of Pkd1 (encoding polycystin-1) affected the expression of key factors involved in the process of ferroptosis, using Western blot and qRT-PCR analysis in Pkd1 mutant renal cells and tissues. We also examined whether treatment with erastin, a ferroptosis inducer, and ferrostain-1, a ferroptosis inhibitor, affected cyst growth in Pkd1 mutant mouse models. RESULTS: We found that kidney cells and tissues lacking Pkd1 exhibit extensive metabolic abnormalities, including reduced expression of the system Xc- amino acid antiporter (critical for import of cystine), of iron exporter (ferroportin), and of GPX4 (a key and negative regulator of ferroptosis). The abnormalities also include increased expression of iron importers (TfR1, DMT1) and HO-1, which in turn result in high iron levels, low GSH and GPX4 activity, increased lipid peroxidation, and propensity to ferroptosis. We further found that erastin increased, and ferrostatin-1 inhibited ferroptotic cell death and proliferation of Pkd1-deficient cells in kidneys from Pkd1 mutant mice. A lipid peroxidation product increased in Pkd1-deficient cells, 4HNE, promoted the proliferation of survived Pkd1 mutant cells via activation of Akt, S6, Stat3, and Rb during the ferroptotic process, contributing to cyst growth. CONCLUSION: These findings indicate that ferroptosis contributes to ADPKD progression and management of ferroptosis may be a novel strategy for ADPKD treatment.


Assuntos
Ferroptose , Rim Policístico Autossômico Dominante/patologia , Animais , Ciclo Celular , Células Cultivadas , Cicloexilaminas/farmacologia , Cicloexilaminas/uso terapêutico , Modelos Animais de Doenças , Progressão da Doença , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Ferro/metabolismo , Peroxidação de Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/genética , Interferência de RNA , Esferoides Celulares , Canais de Cátion TRPP/deficiência , Canais de Cátion TRPP/genética , Transcriptoma
17.
Biochem Pharmacol ; 192: 114716, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34339713

RESUMO

A total number of 18 different isoforms of histone deacetylases (HDACs) which were categorized into 4 classes have been identified in human. HDAC3 is categorized as class I HDACs and is closely related to the occurrence and development of atherosclerosis. Recent evidence has pointed to endothelial-to-mesenchymal transition (EndMT) as a key process in vascular inflammation in atherosclerosis. However, little is known about the effect of HDAC3 on EndMT in atherosclerosis. Therefore, we aimed to investigate the effect of HDAC3 specific inhibitor on EndMT in ApoE-/- mice fed a Western diet and human umbilical vein endothelial cells (HUVECs) induced by inflammatory cytokines. Firstly, we found that HDAC3 expression was up-regulated and EndMT occurred in the aortas of ApoE-/- mice compared with C57BL/6J mice. However, HDAC3 specific inhibitor RGFP966 alleviated atherosclerotic lesions and inhibited EndMT of the atherosclerotic plaque in ApoE-/- mice. Then, in vitro study showed that inflammatory cytokines TNF-α and IL-1ß co-treatment increased the expression of HDAC3 and induced EndMT in HUVECs. HDAC3 inhibition by siRNA or specific inhibitor RGFP966 suppressed EndMT in HUVECs stimulated with TNF-α and IL-1ß. By contrast, HDAC3 overexpression by adenovirus further promoted EndMT of HUVECs. In addition, we found that HDAC3 also regulated the inflammatory response of HUVECs by modulating the expression of inflammatory cytokines and the number of monocytes attached to HUVECs. These above results suggest that HDAC3 inhibitor suppresses EndMT via modulating inflammatory response in ApoE-/- mice and HUVECs.


Assuntos
Aterosclerose/metabolismo , Endotélio/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Mediadores da Inflamação/metabolismo , Acrilamidas/farmacologia , Acrilamidas/uso terapêutico , Animais , Aterosclerose/tratamento farmacológico , Endotélio/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Inibidores de Histona Desacetilases/uso terapêutico , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Células THP-1
18.
J Med Chem ; 64(9): 5816-5837, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33929863

RESUMO

We previously reported that P-retigabine (P-RTG), a retigabine (RTG) analogue bearing a propargyl group at the nitrogen atom in the linker of RTG, displayed moderate anticonvulsant efficacy. Recently, our further efforts led to the discovery of HN37 (pynegabine), which demonstrated satisfactory chemical stability upon deleting the ortho liable -NH2 group and installing two adjacent methyl groups to the carbamate motif. HN37 exhibited enhanced activation potency toward neuronal Kv7 channels and high in vivo efficacy in a range of pre-clinical seizure models, including the maximal electroshock test and a 6 Hz model of pharmacoresistant limbic seizures. With its improved chemical stability, strong efficacy, and better safety margin, HN37 has progressed to clinical trial in China for epilepsy treatment.


Assuntos
Anticonvulsivantes/química , Carbamatos/química , Desenho de Fármacos , Animais , Anticonvulsivantes/uso terapêutico , Carbamatos/metabolismo , Carbamatos/uso terapêutico , Modelos Animais de Doenças , Cães , Avaliação Pré-Clínica de Medicamentos , Estabilidade de Medicamentos , Eletrochoque , Meia-Vida , Humanos , Canais de Potássio KCNQ/química , Canais de Potássio KCNQ/metabolismo , Camundongos , Fenilenodiaminas/química , Fenilenodiaminas/metabolismo , Fenilenodiaminas/uso terapêutico , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/tratamento farmacológico , Convulsões/etiologia , Relação Estrutura-Atividade
19.
Am J Psychiatry ; 178(5): 437-446, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33653118

RESUMO

OBJECTIVE: Preclinical studies point to the KCNQ2/3 potassium channel as a novel target for the treatment of depression and anhedonia, a reduced ability to experience pleasure. The authors conducted the first randomized placebo-controlled trial testing the effect of the KCNQ2/3 positive modulator ezogabine on reward circuit activity and clinical outcomes in patients with depression. METHODS: Depressed individuals (N=45) with elevated levels of anhedonia were assigned to a 5-week treatment period with ezogabine (900 mg/day; N=21) or placebo (N=24). Participants underwent functional MRI during a reward flanker task at baseline and following treatment. Clinical measures of depression and anhedonia were collected at weekly visits. The primary endpoint was the change from baseline to week 5 in ventral striatum activation during reward anticipation. Secondary endpoints included depression and anhedonia severity as measured using the Montgomery-Åsberg Depression Rating Scale (MADRS) and the Snaith-Hamilton Pleasure Scale (SHAPS), respectively. RESULTS: The study did not meet its primary neuroimaging endpoint. Participants in the ezogabine group showed a numerical increase in ventral striatum response to reward anticipation following treatment compared with participants in the placebo group from baseline to week 5. Compared with placebo, ezogabine was associated with a significantly larger improvement in MADRS and SHAPS scores and other clinical endpoints. Ezogabine was well tolerated, and no serious adverse events occurred. CONCLUSIONS: The study did not meet its primary neuroimaging endpoint, although the effect of treatment was significant on several secondary clinical endpoints. In aggregate, the findings may suggest that future studies of the KCNQ2/3 channel as a novel treatment target for depression and anhedonia are warranted.


Assuntos
Anedonia , Carbamatos/uso terapêutico , Transtorno Depressivo Maior/tratamento farmacológico , Canal de Potássio KCNQ2 , Canal de Potássio KCNQ3 , Moduladores de Transporte de Membrana/uso terapêutico , Fenilenodiaminas/uso terapêutico , Recompensa , Estriado Ventral/diagnóstico por imagem , Adulto , Transtorno Depressivo/diagnóstico por imagem , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/fisiopatologia , Transtorno Depressivo Maior/diagnóstico por imagem , Transtorno Depressivo Maior/fisiopatologia , Método Duplo-Cego , Feminino , Neuroimagem Funcional , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Estriado Ventral/fisiopatologia
20.
Int Immunopharmacol ; 93: 107400, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33529911

RESUMO

RGFP966 is a selective inhibitor of histone deacetylase 3 (HDAC3) playing crucial roles in triggering allergic and inflammatory responses. Whereas, its role in allergic rhinitis (AR) remains uncertain. This study sought to illustrate the role and mechanism of HDAC3 inhibitor RGFP966 on allergic and inflammatory responses in murine AR. RGFP966 administration was applied on murine AR. HE staining, PAS staining, toluidine blue staining, immunohistochemistry staining and real-time PCR methods were used to assess eosinophils, goblet cells, mast cells, HDAC3 positive cells and mRNA levels in nasal tissues of mice. HDAC3 activities in nasal tissues were quantified with HDAC3 Activity Assay Kit. We collected blood and nasal lavage fluid (NLF) of mice for assaying IgE, inflammatory cytokines and inflammatory cells. Results indicated that RGFP966 intervention attenuated sneezing, nose rubbing, IgE, inflammatory cytokines, eosinophils, goblet cells, mast cells, inflammatory cells, HDAC3 levles and activities in RGFP966 treated mice. In conclusion, RGFP966 might reduce HDAC3 expression and HDAC3 activities, and then eosinophils and mast cells recruitment, goblet cells proliferation and inflammatory cytokines levels are decreased, resulting in the alleviation of allergic and inflammatory responses in AR mice.


Assuntos
Acrilamidas/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Hipersensibilidade/tratamento farmacológico , Inflamação/tratamento farmacológico , Mucosa Nasal/imunologia , Fenilenodiaminas/uso terapêutico , Alérgenos/imunologia , Animais , Modelos Animais de Doenças , Feminino , Histona Desacetilases/metabolismo , Humanos , Imunoglobulina E/sangue , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...