Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Gan To Kagaku Ryoho ; 50(9): 993-996, 2023 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-37800295

RESUMEN

The implantation of a totally implantable central venous(CV)access port is considered a risk factor for venous thromboembolism( VTE). In the treatment of catheter-related thrombosis(CRT), both European and American guidelines recommend anticoagulation therapy with catheters in place. We experienced 2 cases of upper extremity deep vein thrombosis (UEDVT)after the implantation of CV access ports through the left subclavian vein for adjuvant chemotherapy in patients with resected breast cancer. Both patients were successfully treated with direct oral anticoagulants(DOAC) while the port remained in place with a careful follow-up that included monitoring of serum D-dimer levels. The administration of DOAC to CRT that develops in patients undergoing postoperative adjuvant chemotherapy for breast cancer may be relatively safe, with a low potential for adverse events such as bleeding.


Asunto(s)
Neoplasias de la Mama , Catéteres Venosos Centrales , Trombosis Venosa Profunda de la Extremidad Superior , Tromboembolia Venosa , Trombosis de la Vena , Humanos , Femenino , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/etiología , Trombosis Venosa Profunda de la Extremidad Superior/tratamiento farmacológico , Trombosis Venosa Profunda de la Extremidad Superior/etiología , Catéteres Venosos Centrales/efectos adversos , Tromboembolia Venosa/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Anticoagulantes/efectos adversos
2.
Gan To Kagaku Ryoho ; 50(12): 1339-1341, 2023 Dec.
Artículo en Japonés | MEDLINE | ID: mdl-38247077

RESUMEN

An 86-year-old woman was referred to our hospital after an incidental CT scan of the trunk revealed a mass in the left breast and enlarged axillary lymph nodes. A core needle biopsy(CNB)from a 2 cm mass in the left breast revealed invasive ductal carcinoma, weakly positive result for ER, negative result for PgR, and negative result for HER2. She also had multiple enlarged left supraclavicular lymph nodes and was T2N3cM0, Stage ⅢC on pretreatment evaluation. She was given the S-1 oral drug of choice, starting with 80 mg/day/4-week dosing with a 2-week rest. Eight months after the start of S-1, a partial mastectomy and sentinel lymph node biopsy were performed. Pathological findings showed a pathological complete response(ypTis/ypN0)with only a 2 mm non-invasive carcinoma remnant in the left mammary gland. S-1 is weakly recommended as primary chemotherapy for HER2-negative metastatic recurrent breast cancer, but there are no reports to date of complete response in resection cases. S-1 may be administered to patients with locally advanced breast cancer who cannot tolerate standard drug therapy and may be converted to resection after a successful response.


Asunto(s)
Neoplasias de la Mama , Linfadenopatía , Anciano , Femenino , Humanos , Anciano de 80 o más Años , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/cirugía , Mastectomía , Terapia Neoadyuvante , Axila , Respuesta Patológica Completa
3.
Neurobiol Dis ; 156: 105407, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34058347

RESUMEN

Stroke is one of the leading causes of death worldwide, with intracerebral hemorrhage (ICH) being the most lethal subtype. Neuritin (Nrn) is a neurotropic factor that has been reported to have neuroprotective effects in acute brain and spinal cord injury. However, whether Nrn has a protective role in ICH has not been investigated. In this study, ICH was induced in C57BL/6 J mice by injection of collagenase VII, while the overexpression of Nrn in the striatum was induced by an adeno-associated virus serotype 9 (AAV9) vector. We found that compared with GFP-ICH mice, Nrn-ICH mice showed improved performance in the corner, cylinder and forelimb tests after ICH, and showed less weight loss and more rapid weight recovery. Overexpression of Nrn reduced brain lesions, edema, neuronal death and white matter and synaptic integrity dysfunction caused by ICH. Western blot results showed that phosphorylated PERK and ATF4 were significantly inhibited, while phosphorylation of Akt/mammalian target of rapamycin was increased in the Nrn-ICH group, compared with the GFP-ICH group. Whole cell recording from motor neurons indicated that overexpression of Nrn reversed the decrease of spontaneous excitatory postsynaptic currents (sEPSCs) and action potential frequencies induced by ICH. These data show that Nrn improves neurological deficits in mice with ICH by reducing brain lesions and edema, inhibiting neuronal death, and possibly by increasing neuronal connections.


Asunto(s)
Encéfalo/metabolismo , Hemorragia Cerebral/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Recuperación de la Función/fisiología , Adenina/administración & dosificación , Adenina/análogos & derivados , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Hemorragia Cerebral/patología , Dependovirus/genética , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Furanos/administración & dosificación , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Indoles/administración & dosificación , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Técnicas de Cultivo de Órganos , Piridinas/administración & dosificación , Pirimidinas/administración & dosificación , Recuperación de la Función/efectos de los fármacos
4.
J Neurochem ; 147(1): 40-57, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29920676

RESUMEN

Neuritin is a neurotrophic factor that is activated by neural activity and neurotrophins. Its major function is to promote neurite growth and branching; however, the underlying mechanisms are not fully understood. To address this issue, this study investigated the effects of neuritin on neurite and spine growth and intracellular Ca2+ concentration in rat cerebellar granule neurons (CGNs). Incubation of CGNs for 24 h with neuritin increased neurite length and spine density; this effect was mimicked by insulin and abolished by inhibiting insulin receptor (IR) or mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (ERK) activity. Calcium imaging and western blot analysis revealed that neuritin enhanced the increase in intracellular Ca2+ level induced by high K+ , and stimulated the cell surface expression of CaV 1.2 and CaV 1.3 α subunits of the L-type calcium channel, which was suppressed by inhibition of IR or mitogen-activated protein kinase kinase/ERK. Treatment with inhibitors of L-type calcium channels, calmodulin, and calcineurin (CaN) abrogated the effects of neuritin on neurite length and spine density. A similar result was obtained by silencing nuclear factor of activated T cells c4, which is known to be activated by neuritin in CGNs. These results indicate that IR and ERK signaling as well as the Ca2+ /CaN/nuclear factor of activated T cells c4 axis mediate the effects of neuritin on neurite and spine growth in CGNs. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ Cover Image for this issue: doi: 10.1111/jnc.14195.


Asunto(s)
Canales de Calcio Tipo L/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Cerebelo/citología , Espinas Dendríticas/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuropéptidos/farmacología , Animales , Canales de Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Cerebelo/efectos de los fármacos , Cerebelo/crecimiento & desarrollo , Gránulos Citoplasmáticos/efectos de los fármacos , Femenino , Proteínas Ligadas a GPI/farmacología , Silenciador del Gen , Humanos , Insulina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , Ratas , Ratas Sprague-Dawley , Receptor de Insulina/antagonistas & inhibidores
5.
Acta Pharmacol Sin ; 39(9): 1414-1420, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29595190

RESUMEN

Neuritin is a member of the neurotrophic factor family, which is activated by neural activity and neurotrophins, and promotes neurite growth and branching. It has shown to play an important role in neuronal plasticity and regeneration. It is also involved in other biological processes such as angiogenesis, tumorigenesis and immunomodulation. Thus far, however, the primary mechanisms of neuritin, including whether or not it acts through a receptor or which downstream signals might be activated following binding, are not fully understood. Recent evidence suggests that neuritin may be a potential therapeutic target in several neurodegenerative diseases. This review focuses on the recent advances in studies regarding the newly identified functions of neuritin and the signaling pathways related to these functions. We also discuss current hot topics and difficulties in neuritin research.


Asunto(s)
Neuropéptidos/fisiología , Transducción de Señal/fisiología , Animales , Proteínas Ligadas a GPI/fisiología , Humanos , Trastornos Mentales/etiología , Trastornos Mentales/fisiopatología , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Sinapsis/fisiología
6.
Cereb Cortex ; 27(7): 3842-3855, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28475719

RESUMEN

Neuritin is a neurotrophic factor involved in neural development and synaptic plasticity. However, its role in modulating synaptic transmission remains unclear. Here, we investigated the effects of neuritin on miniature excitatory postsynaptic currents (mEPSCs) and glutamate release in the medial prefrontal cortex (mPFC) in mice. Incubation of mPFC slices with neuritin for 45 min significantly increased mEPSC frequency and glutamate release as measured by high-performance liquid chromatography, which was mimicked by insulin and abrogated by an insulin receptor (IR) inhibitor. Neuritin-induced upregulation of synaptic transmission was correlated with activation of ERK, and inhibition of mitogen-activated protein kinases/extracellular signal-regulated kinases (MEK/ERK) activity attenuated the neuritin-induced increase in mEPSC frequency and glutamate release. T-type calcium channel inhibitors but not the L-type inhibitor abolished the inward calcium current and the effects of neuritin on mEPSC frequency and glutamate release. Western blotting of membrane proteins showed that neuritin promoted surface expression of CaV3.3 α-subunit, which was also eliminated by inhibition of IR or MEK/ERK activity. The effects of neuritin on mEPSC frequency, glutamate release, and CaV3.3 α-subunit expression were inhibited by an intracellular protein-transport inhibitor. These results confirm involvement of the IR and ERK signaling pathway, and provide novel insights into the mechanisms of neuritin function in synaptic transmission.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuropéptidos/farmacología , Corteza Prefrontal/citología , Transmisión Sináptica/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Bloqueadores de los Canales de Calcio/farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Femenino , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/farmacología , Ácido Glutámico/metabolismo , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal , Neuropéptidos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tinción con Nitrato de Plata , Transmisión Sináptica/fisiología , Factores de Tiempo , Transducción Genética
7.
Tohoku J Exp Med ; 245(1): 13-19, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29731479

RESUMEN

Positron emission mammography (PEM) has higher detection sensitivity for breast cancer compared with whole-body positron emission tomography (PET) due to higher spatial resolution. We have developed a new PEM device with high resolution over a wide field of view. This PEM device comprises novel scintillation crystals, praseodymium-doped lutetium aluminum garnet (Pr:LuAG). In the present study, the clinical use of the newly developed PEM for the detection of small breast cancer was compared with that of the conventional PET-computed tomography (PET/CT). Eighty-two patients with breast cancer less than 20 mm (UICC T1) participated in this study, including 23 patients with T1a or T1b breast cancer (less than 10 mm). Histologically-proved lesions were examined by PET/CT and PEM on the same day after injection of [18F]fluoro-2-deoxy-2-fluoro-D-glucose ([18F]FDG), a marker of glycolytic activity. The newly developed PEM showed better sensitivity of cancer detection compared with PET/CT especially in case of the small T1a or T1b lesions. Moreover, when the conventional PET/CT and new PEM were combined, the detection sensitivity with [18F]FDG molecular imaging for T1 (N = 82) and T1a plus T1b breast cancer (N = 23) were 90% and 70%, respectively. The uptake of [18F]FDG was proportional to the histological malignancy of breast cancer. Using the newly-developed PEM with [18F]FDG, we are able to identify and characterize exactly the small breast tumors less than 10 mm in combination with the conventional PET/CT. These data indicate that PEM and PET/CT are synergic and complementary for the detection of small breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/diagnóstico , Mamografía , Tomografía de Emisión de Positrones , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/diagnóstico , Carcinoma Ductal de Mama/diagnóstico por imagen , Femenino , Fluorodesoxiglucosa F18/farmacocinética , Humanos , Procesamiento de Imagen Asistido por Computador , Persona de Mediana Edad , Tomografía Computarizada por Tomografía de Emisión de Positrones
8.
J Biol Chem ; 291(33): 17369-81, 2016 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-27307045

RESUMEN

Neuritin is an important neurotrophin that regulates neural development, synaptic plasticity, and neuronal survival. Elucidating the downstream molecular signaling is important for potential therapeutic applications of neuritin in neuronal dysfunctions. We previously showed that neuritin up-regulates transient potassium outward current (IA) subunit Kv4.2 expression and increases IA densities, in part by activating the insulin receptor signaling pathway. Molecular mechanisms of neuritin-induced Kv4.2 expression remain elusive. Here, we report that the Ca(2+)/calcineurin (CaN)/nuclear factor of activated T-cells (NFAT) c4 axis is required for neuritin-induced Kv4.2 transcriptional expression and potentiation of IA densities in cerebellum granule neurons. We found that neuritin elevates intracellular Ca(2+) and increases Kv4.2 expression and IA densities; this effect was sensitive to CaN inhibition and was eliminated in Nfatc4(-/-) mice but not in Nfatc2(-/-) mice. Stimulation with neuritin significantly increased nuclear accumulation of NFATc4 in cerebellum granule cells and HeLa cells, which expressed IR. Furthermore, NFATc4 was recruited to the Kv4.2 gene promoter loci detected by luciferase reporter and chromatin immunoprecipitation assays. More importantly, data obtained from cortical neurons following adeno-associated virus-mediated overexpression of neuritin indicated that reduced neuronal excitability and increased formation of dendritic spines were abrogated in the Nfatc4(-/-) mice. Together, these data demonstrate an indispensable role for the CaN/NFATc4 signaling pathway in neuritin-regulated neuronal functions.


Asunto(s)
Calcineurina/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Espinas Dendríticas/metabolismo , Regulación de la Expresión Génica/fisiología , Factores de Transcripción NFATC/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Canales de Potasio Shal/biosíntesis , Animales , Calcineurina/genética , Cerebelo/metabolismo , Espinas Dendríticas/genética , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células HeLa , Humanos , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/genética , Proteínas del Tejido Nervioso/genética , Neuropéptidos/genética , Canales de Potasio Shal/genética
9.
Biochem J ; 473(13): 1895-904, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27114559

RESUMEN

GDF-15 (growth/differentiation factor 15) is a novel member of the TGF (transforming growth factor)-ß superfamily that has critical roles in the central and peripheral nervous systems. We reported previously that GDF-15 increased delayed rectifier outward K(+) currents and Kv2.1 α subunit expression through TßRII (TGF-ß receptor II) to activate Src kinase and Akt/mTOR (mammalian target of rapamycin) signalling in rat CGNs (cerebellar granule neurons). In the present study, we found that treatment of CGNs with GDF-15 for 24 h increased the intracellular Ca(2+) concentration ([Ca(2+)]i) in response to membrane depolarization, as determined by Ca(2+) imaging. Whole-cell current recordings indicated that GDF-15 increased the inward Ca(2+) current (ICa) without altering steady-state activation of Ca(2+) channels. Treatment with nifedipine, an inhibitor of L-type Ca(2+) channels, abrogated GDF-15-induced increases in [Ca(2+)]i and ICa The GDF-15-induced increase in ICa was mediated via up-regulation of the Cav1.3 α subunit, which was attenuated by inhibiting Akt/mTOR and ERK (extracellular-signal-regulated kinase) pathways and by pharmacological inhibition of Src-mediated TßRII phosphorylation. Given that Cav1.3 is not only a channel for Ca(2+) influx, but also a transcriptional regulator, our data confirm that GDF-15 induces protein expression via TßRII and activation of a non-Smad pathway, and provide novel insight into the mechanism of GDF-15 function in neurons.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Cerebelo/citología , Factor 15 de Diferenciación de Crecimiento/farmacología , Neuronas/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neuronas/efectos de los fármacos , Nifedipino/farmacología , Proteína Oncogénica v-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
10.
Sheng Li Xue Bao ; 69(1): 109-121, 2017 Feb 25.
Artículo en Zh | MEDLINE | ID: mdl-28217814

RESUMEN

Growth differentiation factor-15 (GDF-15) is a member of the transforming growth factor beta superfamily. GDF-15 expression is dramatically upregulated during acute brain injury, cancer, cardiovascular disease, and inflammation, suggesting its potential value as a disease biomarker. It has been suggested that GDF-15 has neurotropic effects in the nervous system. Our studies showed that GDF-15 modulated the expression of neuronal K+ and Ca2+ ion channels and increased the release of excitatory transmitter in the medial prefrontal cortex of mice. GDF-15 is also involved in the complex modulation of cancer and cardiovascular disease. Here, we reviewed studies involving the modulation of GDF-15 expression and its mechanisms, the primary pathological and physiological functions of GDF-15 in neurological and cardiovascular systems, and its role in cancer progression. The biological effects and the values of GDF-15 in basic research and clinical applications were also addressed.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Factor 15 de Diferenciación de Crecimiento/metabolismo , Neoplasias/fisiopatología , Sistema Nervioso/metabolismo , Animales , Lesiones Encefálicas/fisiopatología , Canales de Calcio/metabolismo , Progresión de la Enfermedad , Humanos , Inflamación , Ratones , Canales de Potasio/metabolismo , Corteza Prefrontal/metabolismo , Factor de Crecimiento Transformador beta , Regulación hacia Arriba
11.
J Cell Mol Med ; 19(10): 2413-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26176998

RESUMEN

Previous work from both our lab and others have indicated that exposure to 50 Hz magnetic fields (ELF-MF) was able to modify ion channel functions. However, very few studies have investigated the effects of MF on γ-aminobutyric acid (GABA) type A receptors (GABA(A) Rs) channel functioning, which are fundamental to overall neuronal excitability. Here, our major goal is to reveal the potential effects of ELF-MF on GABA(A) Rs activity in rat cerebellar granule neurons (CGNs). Our results indicated that exposing CGNs to 1 mT ELF-MF for 60 min. significantly increased GABA(A) R currents without modifying sensitivity to GABA. However, activation of PKA by db-cAMP failed to do so, but led to a slight decrease instead. On the other hand, PKC activation or inhibition by PMA or Bis and Docosahexaenoic acid (DHA) mimicked or eliminated the field-induced-increase of GABA(A) R currents. Western blot analysis indicated that the intracellular levels of phosphorylated PKC (pPKC) were significantly elevated after 60 min. of ELF-MF exposure, which was subsequently blocked by application of DHA or EP1 receptor-specific (prostaglandin E receptor 1) antagonist (SC19220), but not by EP2-EP4 receptor-specific antagonists. SC19220 also significantly inhibited the ELF-MF-induced elevation on GABA(A) R currents. Together, these data obviously demonstrated for the first time that neuronal GABA(A) currents are significantly increased by ELF-MF exposure, and also suggest that these effects are mediated via an EP1 receptor-mediated PKC pathway. Future work will focus on a more comprehensive analysis of the physiological and/or pathological consequences of these effects.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Activación del Canal Iónico , Campos Magnéticos , Neuronas/metabolismo , Proteína Quinasa C/metabolismo , Receptores de GABA-A/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Modelos Biológicos , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Análisis de Regresión , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología
12.
Cell Physiol Biochem ; 37(5): 1903-13, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26584289

RESUMEN

BACKGROUND/AIMS: Arachidonic acid (AA) and its metabolites are important endogenous lipid messengers. In this study, we test the effect of Leukotriene B4 (LTB4), a 5-lipoxygenase metabolite of AA, on L-type calcium channels in A7r5 rat aortic vascular smooth muscle cells. METHODS: L-type calcium channel currents were recorded by a patch-clamp technique. The mRNA expression of CaV1.2 was determined by Real-time RT-PCR. The protein expression of CaV1.2 and p38 activity was determined by Western blot analysis. RESULTS: LTB4 inhibits L-type channel currents in A7r5 cells in a dose-and time- dependent manner. LTB4 reduced the mRNA/protein expression of CaV1.2 channels in A7r5 cells. BLT1 receptor antagonist LY29311 abrogated the inhibitory effect of LTB4, while BLT2 receptor antagonist LY255283 had no effect. 5Z-7-oxozeaenol and SB203580, which block TAK1 and p38 kinase respectively, abrogated the LTB4 inhibitory effect on L-type calcium channels. LTB4 increased p38 activity in A7r5 cells. Blockage of Src, PI3K, JNK and NF-x03BA;B kinase had no effects on LTB4 inhibition of L-type calcium channel currents in A7r5 cells. CONCLUSION: We conclude that LTB4 inhibits L-type calcium channels through BLT1-TAk1-p38 signaling pathway. The LTB4 inhibitory effect on L-type calcium channels may be involved in its pathological processes such as atherosclerosis.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Leucotrieno B4/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Benzoatos/farmacología , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Imidazoles/farmacología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Técnicas de Placa-Clamp , Piridinas/farmacología , ARN Mensajero/metabolismo , Ratas , Receptores de Leucotrieno B4/antagonistas & inhibidores , Receptores de Leucotrieno B4/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
13.
Cell Physiol Biochem ; 36(5): 1699-711, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26183394

RESUMEN

AIMS: PGE2 is one of the most abundant prostanoids in mammalian tissues, but its effect on neuronal receptors has not been well investigated. This study examines the effect of PGE2 on GABAA receptor currents in rat cerebellar granule neurons. METHODS: GABAA currents were recorded using a patch-clamp technique. Cell surface and total protein of GABAA ß1/2/3 subunits was carried out by Western blot analysis. RESULTS: Upon incubation of neurons with PGE2 (1 µM) for 60 minutes, GABAA currents were significantly potentiated. This PGE2-driven effect could be blocked by PKC or CaMKII inhibitors as well as EP1 receptor antagonist, and mimicked by PMA or EP1 receptor agonist. Furthermore, Western blot data showed that PGE2 did not increase the total expression level of GABAA receptors, but significantly increased surface levels of GABAA ß1/2/3 subunits after 1 h of treatment. Consistently, both PKC and CaMKII inhibitors were able to reduce PGE2-induced increases in cell surface expression of GABAA receptors. CONCLUSION: Activation of either the PKC or CaMKII pathways by EP1 receptors mediates the PGE2-induced increase in GABAA currents. This suggests that upregulation of postsynaptic GABAA receptors by PGE2 may have profound effects on cerebellar functioning under physiological and pathological conditions.


Asunto(s)
Dinoprostona/fisiología , Receptores de GABA-A/fisiología , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Cerebelo/metabolismo , Gránulos Citoplasmáticos/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
14.
Biochem J ; 460(1): 35-47, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24597762

RESUMEN

GDF15 (growth/differentiation factor 15), a novel member of the TGFß (transforming growth factor ß) superfamily, plays critical roles in the central and peripheral nervous systems, but the signal transduction pathways and receptor subtypes involved are not well understood. In the present paper, we report that GDF15 specifically increases the IK (delayed-rectifier outward K+ current) in rat CGNs (cerebellar granule neurons) in time- and concentration-dependent manners. The GDF15-induced amplification of the IK is mediated by the increased expression and reduced lysosome-dependent degradation of the Kv2.1 protein, the main α-subunit of the IK channel. Exposure of CGNs to GDF15 markedly induced the phosphorylation of ERK (extracellular-signal-regulated kinase), Akt and mTOR (mammalian target of rapamycin), but the GDF15-induced IK densities and increased expression of Kv2.1 were attenuated only by Akt and mTOR, and not ERK, inhibitors. Pharmacological inhibition of the Src-mediated phosphorylation of TGFßR2 (TGFß receptor 2), not TGFßR1, abrogated the effect of GDF15 on IK amplification and Kv2.1 induction. Immunoprecipitation assays showed that GDF15 increased the tyrosine phosphorylation of TGFßRII in the CGN lysate. The results of the present study reveal a novel regulation of Kv2.1 by GDF15 mediated through the TGFßRII-activated Akt/mTOR pathway, which is a previously uncharacterized Smad-independent mechanism of GDF15 signalling.


Asunto(s)
Cerebelo/fisiología , Factor 15 de Diferenciación de Crecimiento/fisiología , Proteína Oncogénica v-akt/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Canales de Potasio Shab/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Cerebelo/citología , Humanos , Ratas , Ratas Sprague-Dawley , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transducción de Señal/genética
15.
J Cell Mol Med ; 18(6): 1060-70, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24548607

RESUMEN

Although melatonin (MT) has been reported to protect cells against oxidative damage induced by electromagnetic radiation, few reports have addressed whether there are other protective mechanisms. Here, we investigated the effects of MT on extremely low-frequency electromagnetic field (ELF-EMF)-induced Nav activity in rat cerebellar granule cells (GCs). Exposing cerebellar GCs to ELF-EMF for 60 min. significantly increased the Nav current (INa ) densities by 62.5%. MT (5 µM) inhibited the ELF-EMF-induced INa increase. This inhibitory effect of MT is mimicked by an MT2 receptor agonist and was eliminated by an MT2 receptor antagonist. The Nav channel steady-state activation curve was significantly shifted towards hyperpolarization by ELF-EMF stimulation but remained unchanged by MT in cerebellar GC that were either exposed or not exposed to ELF-EMF. ELF-EMF exposure significantly increased the intracellular levels of phosphorylated PKA in cerebellar GCs, and both MT and IIK-7 did not reduce the ELF-EMF-induced increase in phosphorylated PKA. The inhibitory effects of MT on ELF-EMF-induced Nav activity was greatly reduced by the calmodulin inhibitor KN93. Calcium imaging showed that MT did not increase the basal intracellular Ca(2+) level, but it significantly elevated the intracellular Ca(2+) level evoked by the high K(+) stimulation in cerebellar GC that were either exposed or not exposed to ELF-EMF. In the presence of ruthenium red, a ryanodine-sensitive receptor blocker, the MT-induced increase in intracellular calcium levels was reduced. Our data show for the first time that MT protects against neuronal INa that result from ELF-EMF exposure through Ca(2+) influx-induced Ca(2+) release.


Asunto(s)
Calcio/metabolismo , Cerebelo/citología , Gránulos Citoplasmáticos/metabolismo , Campos Electromagnéticos/efectos adversos , Melatonina/farmacología , Sustancias Protectoras/farmacología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Antioxidantes/farmacología , Células Cultivadas , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/efectos de la radiación , Gránulos Citoplasmáticos/efectos de los fármacos , Gránulos Citoplasmáticos/efectos de la radiación , Masculino , Ratones , Oxidación-Reducción , Técnicas de Placa-Clamp , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
16.
J Neurochem ; 128(3): 350-62, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24118019

RESUMEN

In addition to their neurotoxic role in Alzheimer's disease (AD), ß-amyloid peptides (Aßs) are also known to play physiological roles. Here, we show that recombinant Aß40 significantly increased the outward current of the GABA(A) receptor containing (GABA(A)α6) in rat cerebellar granule neurons (CGNs). The Aß40-mediated increase in GABA(A)α6 current was mediated by an increase in GABA(A)α6 protein expression at the translational rather than the transcriptional level. The exposure of CGNs to Aß40 markedly induced the phosphorylation of ERK (pERK) and mammalian target of rapamycin (pmTOR). The increase in GABA(A)α6 current and expression was attenuated by specific inhibitors of ERK or mTOR, suggesting that the ERK and mTOR signaling pathways are required for the effect of Aß40 on GABA(A)α6 current and expression in CGNs. A pharmacological blockade of the p75 neurotrophin receptor (p75(NTR)), but not the insulin or α7-nAChR receptors, abrogated the effect of Aß40 on GABA(A)α6 protein expression and current. Furthermore, the expression of GABA(A)α6 was lower in CGNs from APP(-/-) mice than in CGNs from wild-type mice. Moreover, the internal granule layer (IGL) in APP(-/-) mice was thinner than the IGL in wild-type mice. The injection of Aß40 into the cerebellum reversed this effect, and the application of p75(NTR) blocking antibody abolished the effects of Aß40 on cerebellum morphology in APP(-/-) mice. Our results suggest that low concentrations of Aß40 play a role in regulating CGN maturation through p75(NTR).


Asunto(s)
Péptidos beta-Amiloides/farmacología , Cerebelo/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Receptores de GABA-A/biosíntesis , Serina-Treonina Quinasas TOR/efectos de los fármacos , Precursor de Proteína beta-Amiloide/genética , Animales , Biotinilación , Western Blotting , Cerebelo/citología , Cerebelo/efectos de los fármacos , Femenino , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología
17.
Am J Physiol Cell Physiol ; 305(2): C197-206, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23703525

RESUMEN

Neuregulin-1 (NRG-1) is a member of a family of neurotrophic factors that is required for the differentiation, migration, and development of neurons. NRG-1 signaling is thought to contribute to both neuronal development and the neuropathology of schizophrenia, which is believed to be a neurodevelopmental disorder. However, few studies have investigated the role of NRG-1 on voltage-gated ion channels. In this study, we report that NRG-1 specifically increases the density of transient outward K(+) currents (IA) in rat cerebellar granule neurons (CGNs) in a time-dependent manner without modifying the activation or inactivation properties of IA channels. The increase in IA density is mediated by increased protein expression of Kv4.2, the main α-subunit of the IA channel, most likely by upregulation of translation. The effect of NRG-1 on IA density and Kv4.2 expression was only significant in immature neurons. Mechanistically, both Akt and mammalian target of rapamycin (mTOR) signaling pathways are required for the increased NRG-1-induced IA density and expression of Kv4.2. Moreover, pharmacological blockade of the ErbB4 receptor reduced the effect of NRG-1 on IA density and Kv4.2 induction. Our data reveal, for the first time, that stimulation of ErbB4 signaling by NRG-1 upregulates the expression of K(+) channel proteins via activation of the Akt/mTOR signaling pathway and plays an important role in neuronal development and maturation. NRG1 does not acutely change IA and delayed-rectifier outward (IK) of rat CGNs, suggesting that it may not alter excitability of immature neurons by altering potassium channel property.


Asunto(s)
Receptores ErbB/metabolismo , Neurregulina-1/metabolismo , Potasio/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canales de Potasio Shal/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Movimiento Celular , Receptores ErbB/genética , Regulación de la Expresión Génica/fisiología , Potenciales de la Membrana , Neurregulina-1/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor ErbB-4 , Canales de Potasio Shal/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
18.
Am J Physiol Cell Physiol ; 305(5): C547-57, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804203

RESUMEN

Resveratrol (REV) is a naturally occurring phytoalexin that inhibits neuronal K⁺ channels; however, the molecular mechanisms behind the effects of REV and the relevant α-subunit are not well defined. With the use of patch-clamp technique, cultured cerebellar granule cells, and HEK-293 cells transfected with the K(v)2.1 and K(v)2.2 α-subunits, we investigated the effect of REV on K(v)2.1 and K(v)2.2 α-subunits. Our data demonstrated that REV significantly suppressed Kv2.2 but not Kv2.1 currents with a fast, reversible, and mildly concentration-dependent manner and shifted the activation or inactivation curve of Kv2.2 channels. Activating or inhibiting the cAMP/PKA pathway did not abolish the inhibition of K(v)2.2 current by REV. In contrast, activation of PKC with phorbol 12-myristate 13-acetate mimicked the inhibitory effect of REV on K(v)2.2 by modifying the activation or inactivation properties of Kv2.2 channels and eliminated any further inhibition by REV. PKC and PKC-α inhibitor completely eliminated the REV-induced inhibition of K(v)2.2. Moreover, the effect of REV on K(v)2.2 was reduced by preincubation with antagonists of GPR30 receptor and shRNA for GPR30 receptor. Western blotting results indicated that the levels of PKC-α and PKC-ß were significantly increased in response to REV application. Our data reveal, for the first time, that REV inhibited K(v)2.2 currents through PKC-dependent pathways and a nongenomic action of the oestrogen receptor GPR30.


Asunto(s)
Antioxidantes/farmacología , Neuronas/efectos de los fármacos , Proteína Quinasa C/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Canales de Potasio Shab/antagonistas & inhibidores , Estilbenos/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Neuronas/citología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Cultivo Primario de Células , Proteína Quinasa C/genética , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Resveratrol , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología
19.
J Biol Chem ; 287(49): 41534-45, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23066017

RESUMEN

Neuritin is a new neurotrophic factor discovered in a screen to identify genes involved in activity-dependent synaptic plasticity. Neuritin also plays multiple roles in the process of neural development and synaptic plasticity. The receptors for binding neuritin and its downstream signaling effectors, however, remain unclear. Here, we report that neuritin specifically increases the densities of transient outward K(+) currents (I(A)) in rat cerebellar granule neurons (CGNs) in a time- and concentration-dependent manner. Neuritin-induced amplification of I(A) is mediated by increased mRNA and protein expression of Kv4.2, the main α-subunit of I(A). Exposure of CGNs to neuritin markedly induces phosphorylation of ERK (pERK), Akt (pAkt), and mammalian target of rapamycin (pmTOR). Neuritin-induced I(A) and increased expression of Kv4.2 are attenuated by ERK, Akt, or mTOR inhibitors. Unexpectedly, pharmacological blockade of insulin receptor, but not the insulin-like growth factor 1 receptor, abrogates the effect of neuritin on I(A) amplification and Kv4.2 induction. Indeed, neuritin activates downstream signaling effectors of the insulin receptor in CGNs and HeLa. Our data reveal, for the first time, an unanticipated role of the insulin receptor in previously unrecognized neuritin-mediated signaling.


Asunto(s)
Cerebelo/metabolismo , Regulación de la Expresión Génica , Neuronas/metabolismo , Neuropéptidos/metabolismo , Receptor de Insulina/metabolismo , Canales de Potasio Shal/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Ligadas a GPI/metabolismo , Células HeLa , Humanos , Cinética , Modelos Biológicos , Técnicas de Placa-Clamp , Ratas , Regulación hacia Arriba
20.
Sheng Li Xue Bao ; 65(5): 483-8, 2013 Oct 25.
Artículo en Zh | MEDLINE | ID: mdl-24129728

RESUMEN

Neuritin is a new member of the neurotrophic factor family, whose gene is named cpg15 (candidate plasticity-related gene 15) and can be activated by neural activity or neurotrophins (NTs). Experiments show that neuritin is able to promote the growth and branching of neurites, and plays an important role in neuronal plasticity and neuronal regeneration. Recent studies have proved that neuritin is not only involved in the regulation of various physiological functions in the nervous system, but also related in angiogenesis and tumorigenesis. Here we review the mechanisms involved in cpg15 expression and regulation, biological effects of neuritin, and how neuritin plays its biological activities. The hot issues and difficulties in the study of neuritin are also discussed.


Asunto(s)
Neuritas/fisiología , Plasticidad Neuronal , Neuropéptidos/fisiología , Proteínas Ligadas a GPI/fisiología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA