Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Pharmacol Sci ; 153(1): 1-11, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37524448

RESUMEN

Complex regional pain syndrome (CRPS) is an intractable chronic pain syndrome with various signs and symptoms including allodynia/hyperalgesia, edema, swelling, and skin abnormalities. However, a definitive therapeutic treatment for CRPS has not been established. In CRPS patients, inflammatory cytokines such as TNF-α and IL-1ß have been shown to increase in affected areas, suggesting that these molecules may be potential therapeutic targets for CRPS. Here, we first created a novel CRPS mouse model (CRPS-II-like) via sciatic nerve injury and cast immobilization, which was characterized by mechanical allodynia, local edema, and skin abnormalities, to evaluate the pathophysiology and pharmacotherapy of CRPS. When an anti-TNF-α antibody was consecutively administered near the injured sciatic nerve of CRPS model mice, persistent allodynia and CRPS-related signs in the ipsilateral hindpaw were markedly attenuated to control levels. Perineural administration of anti-TNF-α antibody also suppressed the upregulation of inflammatory cytokines as well as the activation of macrophages and Schwann cells in the injured sciatic nerve. These findings indicate that persistent allodynia and CRPS-related signs in CRPS models are primarily associated with TNF-α-mediated immune responses in injured peripheral nerves, suggesting that perineural treatment with anti-TNF-α antibody might be therapeutically useful.


Asunto(s)
Síndromes de Dolor Regional Complejo , Hiperalgesia , Ratas , Ratones , Animales , Hiperalgesia/tratamiento farmacológico , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Factor de Necrosis Tumoral alfa , Ratas Sprague-Dawley , Síndromes de Dolor Regional Complejo/tratamiento farmacológico , Citocinas , Edema/tratamiento farmacológico , Modelos Animales de Enfermedad
2.
J Pharmacol Sci ; 151(2): 128-133, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36707178

RESUMEN

The Sigma-1 receptor (Sigmar1) is downregulated in heart failure model mice with mitochondrial dysfunction. However, the mechanism in detail has not been investigated. In this study, we investigated the role of Sigmar1 in ER-mitochondria proximity using Sigmar1-knockdown or -overexpressed neonatal rat ventricular myocytes (NRVMs). The endothelin-1 (ET-1)-induced cardiomyocyte hypertrophy was aggravated with the dysregulation of mitochondrial function and ER-mitochondrial junctional formation in Sigmar1-knockdown NRVMs, whereas improved in Sigmar1 overexpressed NRVMs. Our data suggests that the reduction of the cardiac Sigmar1 results in decrease mitochondrial Ca2+ influx and promotes mitochondrial fission, followed by reduced ER-mitochondria proximity, exacerbating ET-1-induced cardiomyocyte injury.


Asunto(s)
Insuficiencia Cardíaca , Receptores sigma , Animales , Ratones , Ratas , Homeostasis/genética , Mitocondrias , Miocitos Cardíacos/metabolismo , Receptores sigma/genética , Receptores sigma/metabolismo , Retículo Endoplásmico/metabolismo , Calcio/metabolismo , Receptor Sigma-1
3.
Front Psychiatry ; 13: 859198, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35432011

RESUMEN

Autism spectrum disorder (ASD) is a heterogeneous disorder characterized by repetitive behaviors and social impairments, often accompanied by learning disabilities. It has been documented that the neuropeptide oxytocin (OXT) ameliorates core symptoms in patients with ASD. We recently reported that chronic administration of intranasal OXT reversed social and learning impairments in prenatally valproic acid (VPA)-exposed rats. However, the underlying molecular mechanisms remain unclear. Here, we explored molecular alterations in the hippocampus of rats and the effects of chronic administration of intranasal OXT (12 µg/kg/d). Microarray analyses revealed that prenatal VPA exposure altered gene expression, a part of which is suggested as a candidate in ASD and is involved in key features including memory, developmental processes, and epilepsy. OXT partly improved the expression of these genes, which were predicted to interact with those involved in social behaviors and hippocampal-dependent memory. Collectively, the present study documented molecular profiling in the hippocampus related to ASD and improvement by chronic treatment with OXT.

4.
Acta Pharmacol Sin ; 43(3): 552-562, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33935286

RESUMEN

We previously show that fatty acid-binding protein 3 (FABP3) triggers α-synuclein (Syn) accumulation and induces dopamine neuronal cell death in Parkinson disease mouse model. But the role of fatty acid-binding protein 7 (FABP7) in the brain remains unclear. In this study we investigated whether FABP7 was involved in synucleinopathies. We showed that FABP7 was co-localized and formed a complex with Syn in Syn-transfected U251 human glioblastoma cells, and treatment with arachidonic acid (100 M) significantly promoted FABP7-induced Syn aggregation, which was associated with cell death. We demonstrated that synthetic FABP7 ligand 6 displayed a high affinity against FABP7 with Kd value of 209 nM assessed in 8-anilinonaphthalene-1-sulfonic acid (ANS) assay; ligand 6 improved U251 cell survival via disrupting the FABP7-Syn interaction. We showed that activation of phospholipase A2 (PLA2) by psychosine (10 M) triggered oligomerization of endogenous Syn and FABP7, and induced cell death in both KG-1C human oligodendroglia cells and oligodendrocyte precursor cells (OPCs). FABP7 ligand 6 (1 M) significantly decreased Syn oligomerization and aggregation thereby prevented KG-1C and OPC cell death. This study demonstrates that FABP7 triggers α-synuclein oligomerization through oxidative stress, while FABP7 ligand 6 can inhibit FABP7-induced Syn oligomerization and aggregation, thereby rescuing glial cells and oligodendrocytes from cell death.


Asunto(s)
Proteína de Unión a los Ácidos Grasos 7/metabolismo , Neuroglía/metabolismo , Oligodendroglía/metabolismo , Estrés Oxidativo/fisiología , alfa-Sinucleína/metabolismo , Animales , Ácido Araquidónico/farmacología , Muerte Celular/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Fosfolipasas A2/efectos de los fármacos , Unión Proteica/fisiología , Psicosina/farmacología
5.
J Pharmacol Sci ; 146(1): 1-9, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33858649

RESUMEN

Alzheimer's disease (AD) accounts for the majority of dementia among the elderly. In addition to cognitive impairment, behavioral and psychological symptoms (BPSD) such as depression tendency and increased aggression impose a great burden on the patient. However, there is still no rational therapeutic drug for BPSD. Recently, we developed a novel AD therapeutic candidate, SAK3, and demonstrated that it improved cognitive dysfunction in AppNL-G-F/NL-G-F knock-in (NL-G-F) mice. In this study, we investigated whether acute SAK3 administration improved BPSD in addition to cognitive improvement. Acute SAK3 administration improved BPSD, including anxiolytic and depressive-like behaviors, and ameliorated aggressive behaviors. Furthermore, continuous SAK3 administration improved anxiolytic and depressive-like behaviors. Intriguingly, the anti-anxiolytic and cognitive improvement lasted two weeks after the withdrawal of SAK3, whereas the anti-depressive action did not. Taken together, SAK3 had comprehensive beneficial effects on BPSD behavior.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Conducta Animal/efectos de los fármacos , Canales de Calcio Tipo T/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Imidazoles/administración & dosificación , Imidazoles/farmacología , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/etiología , Compuestos de Espiro/administración & dosificación , Compuestos de Espiro/farmacología , Animales , Ansiolíticos , Antidepresivos , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Masculino , Ratones Endogámicos C57BL
6.
J Biol Chem ; 296: 100663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33862084

RESUMEN

α-synuclein (αSyn) is a protein known to form intracellular aggregates during the manifestation of Parkinson's disease. Previously, it was shown that αSyn aggregation was strongly suppressed in the midbrain region of mice that did not possess the gene encoding the lipid transport protein fatty acid binding protein 3 (FABP3). An interaction between these two proteins was detected in vitro, suggesting that FABP3 may play a role in the aggregation and deposition of αSyn in neurons. To characterize the molecular mechanisms that underlie the interactions between FABP3 and αSyn that modulate the cellular accumulation of the latter, in this report, we used in vitro fluorescence assays combined with fluorescence microscopy, transmission electron microscopy, and quartz crystal microbalance assays to characterize in detail the process and consequences of FABP3-αSyn interaction. We demonstrated that binding of FABP3 to αSyn results in changes in the aggregation mechanism of the latter; specifically, a suppression of fibrillar forms of αSyn and also the production of aggregates with an enhanced cytotoxicity toward mice neuro2A cells. Because this interaction involved the C-terminal sequence region of αSyn, we tested a peptide derived from this region of αSyn (αSynP130-140) as a decoy to prevent the FABP3-αSyn interaction. We observed that the peptide competitively inhibited binding of αSyn to FABP3 in vitro and in cultured cells. We propose that administration of αSynP130-140 might be used to prevent the accumulation of toxic FABP3-αSyn oligomers in cells, thereby preventing the progression of Parkinson's disease.


Asunto(s)
Amiloide/antagonistas & inhibidores , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Neuroblastoma/patología , Fragmentos de Péptidos/farmacología , Agregación Patológica de Proteínas/prevención & control , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Animales , Proteína 3 de Unión a Ácidos Grasos/genética , Humanos , Ratones , Neuroblastoma/genética , Neuroblastoma/metabolismo , Células Tumorales Cultivadas , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/genética
7.
Pharm Res ; 38(3): 479-490, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33646504

RESUMEN

PURPOSE: Fatty acid-binding protein 7 (FABP7) involved in intracellular lipid dynamics, is highly expressed in melanomas and associated with decreased patient survival. Several studies put FABP7 at the center of melanoma cell proliferation. However, the underlying mechanisms are not well deciphered. This study examines the effects of FABP7 on Wnt/ß-catenin signaling that enhances proliferation in melanoma cells. METHODS: Skmel23 cells with FABP7 silencing and Mel2 cells overexpressed with wild-type FABP7 (FABP7wt) and mutated FABP7 (FABP7mut) were used. Cell proliferation and migration were analyzed by proliferation and wound-healing assay, respectively. Transcriptional activation of the Wnt/ß-catenin signaling was measured by luciferase reporter assay. The effects of a specific FABP7 inhibitor, MF6, on proliferation, migration, and modulation of the Wnt/ß-catenin signaling were examined. RESULTS: FABP7 siRNA knockdown in Skmel23 decreased proliferation and migration, cyclin D1 expression, as well as Wnt/ß-catenin activity. Similarly, FABP7wt overexpression in Mel2 cells increased these effects, but FABP7mut abrogated these effects. Pharmacological inhibition of FABP7 function with MF6 suppressed FABP7-regulated proliferation of melanoma cells. CONCLUSION: These results suggest the importance of the interaction between FABP7 and its ligands in melanoma proliferation modulation, and the beneficial implications of therapeutic targeting of FABP7 for melanoma treatment.


Asunto(s)
Proteína de Unión a los Ácidos Grasos 7/metabolismo , Melanoma/metabolismo , Proteínas Supresoras de Tumor/metabolismo , beta Catenina/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Proteína de Unión a los Ácidos Grasos 7/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ligandos , ARN Interferente Pequeño , Proteínas Supresoras de Tumor/genética , Vía de Señalización Wnt , beta Catenina/genética
8.
Biomedicines ; 9(2)2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499263

RESUMEN

The accumulation of α-synuclein (αSyn) has been implicated as a causal factor in the pathogenesis of Parkinson's disease (PD). There is growing evidence that supports mitochondrial dysfunction as a potential primary cause of dopaminergic neuronal death in PD. Here, we focused on reciprocal interactions between αSyn aggregation and mitochondrial injury induced by oxidative stress. We further investigated whether epidermal fatty acid-binding protein 5 (FABP5) is related to αSyn oligomerization/aggregation and subsequent disturbances in mitochondrial function in neuronal cells. In the presence of rotenone, a mitochondrial respiratory chain complex I inhibitor, co-overexpression of FABP5 with αSyn significantly decreased the viability of Neuro-2A cells compared to that of αSyn alone. Under these conditions, FABP5 co-localized with αSyn in the mitochondria, thereby reducing mitochondrial membrane potential. Furthermore, we confirmed that pharmacological inhibition of FABP5 by its ligand prevented αSyn accumulation in mitochondria, which led to cell death rescue. These results suggested that FABP5 is crucial for mitochondrial dysfunction related to αSyn oligomerization/aggregation in the mitochondria induced by oxidative stress in neurons.

9.
Int J Mol Sci ; 21(15)2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32752296

RESUMEN

Recently, we developed the fatty acid-binding protein 3 (FABP3) ligand MF1 (4-(2-(1-(2-chlorophenyl)-5-phenyl-1H-pyrazol-3-yl)phenoxy) butanoic acid) as a therapeutic candidate for α-synucleinopathies. MF1 shows affinity towards γ-aminobutyric acid type-A (GABAA) receptor, but its effect on the receptor remains unclear. Here, we investigate the pharmacological properties of MF1 on the GABAA receptor overexpressed in Neuro2A cells. While MF1 (1-100 µm) alone failed to evoke GABA currents, MF1 (1 µm) promoted GABA currents during GABA exposure (1 and 10 µm). MF1-promoted GABA currents were blocked by flumazenil (10 µm) treatment, suggesting that MF1 enhances receptor function via the benzodiazepine recognition site. Acute and chronic administration of MF1 (0.1, 0.3 and 1.0 mg/kg, p.o.) significantly attenuated status epilepticus (SE) and the mortality rate in pilocarpine (PILO: 300 mg/kg, i.p.)-treated mice, similar to diazepam (DZP: 5.0 mg/kg, i.p.). The anti-epileptic effects of DZP (5.0 mg/kg, i.p.) and MF1 (0.3 mg/kg, p.o.) were completely abolished by flumazenil (25 mg/kg, i.p.) treatment. Pentylenetetrazol (PTZ: 90 mg/kg, i.p.)-induced seizures in mice were suppressed by DZP (5.0 mg/kg, i.p.), but not MF1. Collectively, this suggests that MF1 is a mild enhancer of the GABAA receptor and exercises anti-epileptic effects through the receptor's benzodiazepine recognition site in PILO-induced SE models.


Asunto(s)
Anticonvulsivantes/farmacología , Benzodiazepinas/farmacología , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Receptores de GABA-A/metabolismo , Estado Epiléptico/tratamiento farmacológico , Animales , Anticonvulsivantes/química , Anticonvulsivantes/metabolismo , Benzodiazepinas/metabolismo , Sitios de Unión , Línea Celular Tumoral , Diazepam/metabolismo , Diazepam/farmacología , Flumazenil/metabolismo , Flumazenil/farmacología , Ligandos , Masculino , Ratones Endogámicos ICR , Pentilenotetrazol/metabolismo , Pentilenotetrazol/farmacología , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Estado Epiléptico/metabolismo
10.
Int J Mol Sci ; 21(11)2020 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-32481611

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia and is characterized by neuropathological hallmarks consisting of accumulation of extracellular amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles (NFT). Recently, we have identified a new AD therapeutic candidate, ethyl-8'-methyl-2',4-dioxo-2-(piperidin-1-yl)-2'H-spiro[cyclopentane-1,3'-imidazo [1,2-a] pyridin]-2-ene-3-carboxylate (SAK3), which ameliorates the AD-like pathology in AppNL-F/NL-F knock-in mice. However, the detailed mechanism underlying the therapeutic effects of SAK3 remains unclear. In this study, we found that SAK3 administration improved the reduced proteasome activity through the activation of CaMKII/Rpt6 signaling in AppNL-F/NL-F knock-in (NL-G-F) mice. Moreover, spine abnormalities observed in NL-G-F mice were significantly reversed by SAK3 administration. Along with this, cognitive impairments found in NL-G-F mice were markedly ameliorated by SAK3. In summary, our data suggest that SAK3 administration increases the activity of the proteasome via activation of the CaMKII/Rpt6 signaling pathway, contributing to improvements in spine abnormalities and cognitive deficits in NL-G-F mice. Overall, our findings suggest that SAK3 might be a new attractive drug candidate, representing a new mechanism for the treatment of AD pathology.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Precursor de Proteína beta-Amiloide/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Disfunción Cognitiva/tratamiento farmacológico , Imidazoles/farmacología , Complejo de la Endopetidasa Proteasomal/genética , Columna Vertebral/patología , Compuestos de Espiro/farmacología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Disfunción Cognitiva/metabolismo , Espinas Dendríticas/metabolismo , Femenino , Técnicas de Sustitución del Gen , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Fosforilación , Placa Amiloide/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal
11.
J Pharmacol Sci ; 143(4): 264-271, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32499096

RESUMEN

Fatty acid-binding proteins (FABPs) belong to a family of proteins that transports fatty acids in the cytosol and regulates cellular functions like membrane phospholipid synthesis, lipid metabolism, and mitochondrial ß oxidation. In this study, we synthesized ten novel derivatives from BMS309403, a biphenyl azole compound specific for FABP4, and analyzed their affinity and specificity for FABP3, FABP4, and FABP5, which possess 60% of homology in amino acid sequence. Here, we used 1-anilinonaphthalene 8-sulfonic acid (ANS) displacement assay and found that Ligand 1 has highest affinity for FABP3, with comparable affinity for FABP4 and FABP5. The apparent dissociation constant of BMS309403 was identical to that of arachidonic acid and docosahexaenoic acid. Docking studies with X-ray structural data showed that these novel derivatives obtained by the substitution of phenoxyacetic acid in BMS309403 but not BMS309403 have high or moderate affinity for FABP3. We further found that substitution of a phenyl group and alkyl group caused steric hindrance between 16F, the portal loop and 115L, 117L, respectively, leading to decrease in their affinity for FABPs. In conclusion, our study provides a novel strategy for development of specific ligand for each FABP.


Asunto(s)
Proteínas de Unión a Ácidos Grasos , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica
12.
Pharmacol Biochem Behav ; 191: 172891, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32126223

RESUMEN

The main symptom of Parkinson's disease (PD) is motor dysfunction and remarkably approximately 30-40% of PD patients exhibit cognitive impairments. Recently, we have developed MF8, a heart-type fatty acid-binding protein (FABP3)-specific ligand, which can inhibit α-synuclein (α-syn) oligomerization induced by arachidonic acid in FABP3 overexpressing neuro2A cells. The present study aimed to determine whether MF8 attenuates dopaminergic neuronal death and motor and cognitive impairments in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice model. MF8 can penetrate the blood-brain barrier and its peak brain concentration (21.5 ± 2.1 nM) was achieved 6 h after the oral administration (1.0 mg/kg). We also compared its effects and pharmacological action with those of L-DOPA (3,4-dihydroxy-l-phenylalanine). PD model mice were developed by administering MPTP (25 mg/kg, i.p.) once a day for five consecutive days. Twenty-four hours after the final MPTP injection, mice were administered MF8 (0.3, 1.0 mg/kg, p.o.) or L-DOPA (25 mg/kg, i.p.) once a day for 28 consecutive days and subjected to behavioral and histochemical studies. MF8 (1.0 mg/kg, p.o.), but not L-DOPA, inhibited the dopaminergic neuronal death in the ventral tegmental area and the substantia nigra pars compacta region of the MPTP-treated mice. MF8 also improved both, motor and cognitive functions, while L-DOPA ameliorated only motor dysfunction. Taken together, our results showed that MF8 attenuated the MPTP-induced dopaminergic neuronal death associated with PD pathology. We present MF8 as a novel disease-modifying therapeutic molecule for PD, which acts via a mechanism different from that of L-DOPA.


Asunto(s)
Antiparkinsonianos/administración & dosificación , Muerte Celular/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , Animales , Barrera Hematoencefálica/efectos de los fármacos , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Levodopa/administración & dosificación , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/etiología , Agregación Patológica de Proteínas/tratamiento farmacológico , alfa-Sinucleína/metabolismo
13.
J Biol Chem ; 295(51): 17573-17587, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33453999

RESUMEN

Genetic mutations related to ALS, a progressive neurological disease, have been discovered in the gene encoding σ-1 receptor (σ1R). We previously reported that σ1RE102Q elicits toxicity in cells. The σ1R forms oligomeric states that are regulated by ligands. Nevertheless, little is known about the effect of ALS-related mutations on oligomer formation. Here, we transfected NSC-34 cells, a motor neuronal cell line, and HEK293T cells with σ1R-mCherry (mCh), σ1RE102Q-mCh, or nontagged forms to investigate detergent solubility and subcellular distribution using immunocytochemistry and fluorescence recovery after photobleaching. The oligomeric state was determined using crosslinking procedure. σ1Rs were soluble to detergents, whereas the mutants accumulated in the insoluble fraction. Within the soluble fraction, peak distribution of mutants appeared in higher sucrose density fractions. Mutants formed intracellular aggregates that were co-stained with p62, ubiquitin, and phosphorylated pancreatic eukaryotic translation initiation factor-2-α kinase in NSC-34 cells but not in HEK293T cells. The aggregates had significantly lower recovery in fluorescence recovery after photobleaching. Acute treatment with σ1R agonist SA4503 failed to improve recovery, whereas prolonged treatment for 48 h significantly decreased σ1RE102Q-mCh insolubility and inhibited apoptosis. Whereas σ1R-mCh formed monomers and dimers, σ1RE102Q-mCh also formed trimers and tetramers. SA4503 reduced accumulation of the four types in the insoluble fraction and increased monomers in the soluble fraction. The σ1RE102Q insolubility was diminished by σ1R-mCh co-expression. These results suggest that the agonist and WT σ1R modify the detergent insolubility, toxicity, and oligomeric state of σ1RE102Q, which may lead to promising new treatments for σ1R-related ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Receptores sigma/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Estrés del Retículo Endoplásmico , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mutagénesis Sitio-Dirigida , Piperazinas/farmacología , Agregado de Proteínas/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Receptores sigma/agonistas , Receptores sigma/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Solubilidad , Proteína Fluorescente Roja , Receptor Sigma-1
14.
Mol Neurobiol ; 56(12): 7911-7928, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31129809

RESUMEN

Nicotine in tobacco causes psychological dependence through its rewarding effect in the central nervous system (CNS). Although nicotine dependence is explained by dopamine receptor (DR) signaling together with nicotinic acetylcholine receptors (nAChRs), the synaptic molecular mechanism underlying the interaction between dopamine receptor and nAChRs remains unclear. Since reward signaling is mediated by dopamine receptors, we hypothesized that the dopamine D2 receptor (D2R), in part, mediates the synaptic modulation of nicotine-induced conditioned place preference (CPP) in addition to dopamine D1 receptor. To investigate the involvement of D2R, wild-type (WT) and dopamine D2 receptor knockout (D2RKO) mice were assessed using the CPP task after induction of nicotine-induced CPP. As expected, D2RKO mice failed to induce CPP behaviors after repeated nicotine administration (0.5 mg/kg). When kinase signaling was assessed in the nucleus accumbens and hippocampal CA1 region after repeated nicotine administration, both Ca2+/calmodulin-dependent protein kinase (CaMKII) and extracellular signal-regulated kinase (ERK) were upregulated in WT mice but not in D2RKO mice. Likewise, nicotine-induced CPP was associated with elevation of pro- brain-derived neurotropic factor (BDNF) and BDNF protein levels in WT mice, but not in D2RKO mice. Taken together, in addition to dopamine D1 receptor signaling, dopamine D2 receptor signaling is critical for induction of nicotine-induced CPP in mice.


Asunto(s)
Condicionamiento Psicológico/fisiología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Receptores de Dopamina D2/fisiología , Transducción de Señal/fisiología , Animales , Condicionamiento Psicológico/efectos de los fármacos , Antagonistas de los Receptores de Dopamina D2/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/efectos de los fármacos
15.
J Pharmacol Sci ; 139(4): 319-324, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30871872

RESUMEN

Phospholipids are structural components of cellular membranes that play important roles as precursors for various signaling pathways in modulating neuronal membrane function and maintenance of the intracellular environment. Phosphatidylcholine (PtdCho) is the most abundant cellular phospholipid. Citicoline and docosahexaenoic acid (DHA) are essential intermediates in the synthesis of PtdCho. Both PtdCho intermediates have independently shown neuroprotective effects in cerebral ischemia, but their combined effect is unknown. This study aimed to investigate the combined effect of oral citicoline and DHA treatment on improvement of cognitive deficits following cerebral ischemia using a 20-min bilateral common carotid artery occlusion (BCCAO) mouse model. BCCAO ischemic mice were treated for a total of 11 days with a combination of citicoline (40 mg/kg body weight/day) and DHA (300 mg/kg body weight/day) or each alone. Combined citicoline and DHA synergistically and significantly improved learning and memory ability of ischemic mice compared with either alone. Further, citicoline and DHA treatment significantly prevented neuronal cell death, and slightly increased DHA-containing PtdCho in the hippocampus, albeit not significantly. Taken together, these findings suggest that combined citicoline and DHA treatment may have synergistic benefits for partially improving memory deficits following transient brain ischemia.


Asunto(s)
Isquemia Encefálica/complicaciones , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Citidina Difosfato Colina/administración & dosificación , Citidina Difosfato Colina/farmacología , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/farmacología , Fármacos Neuroprotectores , Animales , Reacción de Prevención/efectos de los fármacos , Región CA1 Hipocampal/patología , Supervivencia Celular , Disfunción Cognitiva/patología , Disfunción Cognitiva/psicología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Neuronas/patología , Reconocimiento en Psicología/efectos de los fármacos , Resultado del Tratamiento
16.
J Pharmacol Sci ; 139(2): 51-58, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30600144

RESUMEN

Low-threshold Ca2+ spikes are mediated by T-type Ca2+ channels, which have electrophysiological properties of fast inactivation and slow deactivation kinetics. A low membrane potential of approximately -60 mV is sufficient to trigger channel opening. We recently introduced a novel T-type Ca2+ channel enhancer that improves cognition and inhibits amyloid beta aggregation in an Alzheimer's disease (AD) mouse model. The enhancer stimulates ACh release, Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, and neurogenesis in the hippocampus. Then, we discuss how T-type Ca2+ channel enhancer improves cognition and impaired neurogenesis and how CaMKII signaling in neurodegenerative diseases reduces amyloid beta aggregation. We provide a perspective of the potential AD therapies to target CaMKII signaling. In this context, we overview our attempts leading to the development of a T-type Ca2+ channel enhancer as cognitive enhancer, the action of which has been associated with CaMKII and presumably proteasome activity.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Canales de Calcio Tipo T/fisiología , Imidazoles/uso terapéutico , Compuestos de Espiro/uso terapéutico , Acetilcolina/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hipocampo/fisiología , Humanos , Imidazoles/farmacología , Neurogénesis , Complejo de la Endopetidasa Proteasomal/metabolismo , Compuestos de Espiro/farmacología
17.
Brain Res ; 1707: 190-197, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30496735

RESUMEN

In Parkinson's disease (PD), α-synuclein (αSyn) accumulation and inclusion triggers dopamine neuronal death and synapse dysfunction in vivo. We previously reported that fatty acid-binding protein 3 (FABP3) is highly expressed in the brain and accelerates αSyn oligomerization when cells are exposed to 1-Methyl-1,2,3,6-tetrahydropiridine (MPTP). Here, we demonstrate that αSyn oligomerization was markedly enhanced by co-overexpressing FABP3 in neuro-2A cells when cells were treated with arachidonic acid (AA). We developed FABP3 ligands, which bind to the fatty acid binding domain of FABP3, using an 8-Anilinonaphthalene-1-sulfonic acid (ANS) assay with a recombinant FABP3 protein. The prototype for the FABP4 ligand, BMS309403, has no affinity for FABP3. We developed more FABP3-specific ligands derived from the chemical structure of BMS309403. Like AA, ligands 1, 7, and 8 had a relatively high affinity for FAPB3 in the ANS assay. Then, we evaluated the inhibition of αSyn oligomerization in neuro-2A cells co-overexpressing FABP3 and αSyn. Importantly, AA treatments markedly enhanced αSyn oligomerization in the co-expressing cells. Ligands 1, 7, and 8 significantly reduced AA-induced αSyn oligomerization in neuro-2A cells. Taken together, our results indicate that FABP3 ligands that target FABP3 may be used as potential therapeutics that inhibit αSyn aggregation in vivo.


Asunto(s)
Proteína 3 de Unión a Ácidos Grasos/metabolismo , Sinucleinopatías/metabolismo , alfa-Sinucleína/metabolismo , Animales , Ácido Araquidónico/farmacología , Encéfalo/metabolismo , Línea Celular Tumoral , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteína 3 de Unión a Ácidos Grasos/fisiología , Ligandos , Ratones , Enfermedad de Parkinson/fisiopatología , Sustancia Negra/metabolismo
18.
Neuroscience ; 399: 89-102, 2019 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-30579831

RESUMEN

KY-226 is a protein tyrosine phosphatase 1B (PTP1B) inhibitor that protects neurons from cerebral ischemic injury. KY-226 restores Akt (protein kinase B) phosphorylation and extracellular signal-regulated kinase (ERK) reduction in transient middle cerebral artery occlusion (tMCAO) damage. However, the mechanisms underlying the neuroprotective effects of KY-226 are unclear. To address this, the effects of KY-226 on blood-brain barrier (BBB) dysfunction were examined in tMCAO mice. KY-226 (10 mg/kg, i.p.) was administered to ICR mice 30 min after 2 h of tMCAO. To assess Akt or ERK involvement, wortmannin (i.c.v.) or U0126 (i.v.), selective inhibitors of PI3K and ERK, respectively, were administered to mice 30 min before ischemia. BBB integrity was assessed by Evans blue leakage 24 h post-reperfusion. The levels of tight junction (TJ) proteins, ZO-1 and occludin, were measured by western blotting; ZO-1 mRNA level was measured by RT-PCR. Compared to vehicle, KY-226 treatment prevented BBB breakdown and reduction in TJ protein levels. KY-226 treatment restored ZO-1 mRNA levels post-reperfusion. Pre-administration of wortmannin or U0126 blocked the protective effects of KY-226 on ZO-1 protein and mRNA reduction in tMCAO mice. In bEnd.3 cells, lipopolysaccharide treatment reduced mRNA and protein levels of ZO-1, an effect rescued by KY-226 treatment. Further, KY-226 treatment restored phosphorylation of pAkt (T308) and its downstream target forkhead box protein O1 (FoxO1) (S256) in bEnd.3 cells. Collectively, we demonstrate that KY-226 protects BBB integrity by restoration of TJ proteins, an effect partly mediated by Akt/FoxO1 pathway activation. Thus, protection of BBB integrity likely underlies KY-226-induced neuroprotection in tMCAO mice.


Asunto(s)
Benzamidas/farmacología , Compuestos de Bifenilo/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Proteína Forkhead Box O1/metabolismo , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Línea Celular , Modelos Animales de Enfermedad , Lipopolisacáridos , Masculino , Ratones Endogámicos ICR , Ocludina/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Proteína de la Zonula Occludens-1/metabolismo
19.
Pharmacology ; 102(3-4): 223-232, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30099467

RESUMEN

Nicotine is a stimulatory component in tobacco that activates the central nervous system reward pathway and causes nicotine dependence. We found that the anti-inflammatory agent, curcuminoid, prevents nicotine dependence and relapse, as assessed by the conditioned placed preference test. Curcuminoid (1, 3.2, and 10 mg·kg-1, oral) dose-dependently inhibited nicotine dependence and enhanced nicotine extinction when administrated 30 min prior to nicotine administration (0.5 mg·kg-1, i.p.) for 7 days. In addition, curcuminoid significantly suppressed the priming effects of nicotine and inhibited acetylcholinesterase (AChE) activity. Taken together, curcuminoid ameliorates nicotine dependence and relapse, in part via the inhibition of the AChE activity in the brain.


Asunto(s)
Acetilcolinesterasa/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Encéfalo/enzimología , Antagonistas Colinérgicos/farmacología , Curcumina/análogos & derivados , Curcumina/farmacología , Tabaquismo/tratamiento farmacológico , Tabaquismo/enzimología , Animales , Encéfalo/efectos de los fármacos , Bupropión/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Tabaquismo/prevención & control
20.
Brain Res ; 1694: 1-12, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29705606

RESUMEN

Akt (Protein kinase B, PKB), a serine/threonine kinase, plays a critical role in cell development, growth, and survival. Akt phosphorylation mediates a neuroprotective effect against ischemic injury. Recently, a protein-tyrosine phosphatase-1B (PTP1B) inhibitor (KY-226) was developed to elicit anti-diabetic and anti-obesity effects via enhancement of insulin signaling. Previously, we reported that the nonselective PTP1B inhibitor, sodium orthovanadate, rescued neurons from delayed neuronal death during brain ischemia. In this study, we confirmed the ameliorative effects of KY-226 on ischemia/reperfusion (I/R) injury using a murine model of middle cerebral artery occlusion (MCAO). ICR mice were subjected to MCAO for 2 h followed by reperfusion. Although KY-226 permeability was poor through the blood-brain barrier (BBB) of normal mice, it could penetrate through the BBB of mice after I/R insult. Intraperitoneal KY-226 administration elicited dose-dependent reductions in infarcted brain areas and improved neurological deficits. The neuroprotective effects of KY-266 were obtained when administered within 0.5 h after reperfusion. KY-226 (10 mg/kg) also restored reduced Akt phosphorylation and eNOS phosphorylation (Ser-1177) levels following I/R insult. Moreover, 10 mg/kg of KY-226 improved I/R-induced decreased extracellular signal-regulated kinase (ERK) phosphorylation. Furthermore, KY-226 attenuated the generation of reactive oxygen species (ROS) in mouse cortex. These results suggest that KY-226 may act as a novel therapeutic candidate for ischemic stroke. Activation of Akt and ERK possibly underlie the neuroprotective mechanism of KY-226.


Asunto(s)
Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Daño por Reperfusión/tratamiento farmacológico , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Muerte Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos ICR , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA