Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Card Surg ; 36(9): 3452-3455, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34216385

RESUMEN

BACKGROUND: Cardiac angiosarcoma is a rare but highly malignant cardiac tumor. It is characterized by poor prognosis, and current treatment approaches are not effective. CASE PRESENTATION: A 37-year-old female with 35 weeks pregnancy experienced chest tightness and shortness of breath for 1 month. She was diagnosed with primary cardiac angiosarcoma. Delivery of fetus was performed early to treat the mother. The patient underwent resection of the tumor then she was treated with chemotherapy. However, the tumor recurred 11 months after surgery. CONCLUSION: Angiosarcoma is a highly malignant tumor explaining recurrence of the tumor recurred after surgery. Cardiac angiosarcoma should be treated through a comprehensive treatment plan, comprising surgery, radiotherapy, and chemotherapy approaches.


Asunto(s)
Neoplasias Cardíacas , Hemangiosarcoma , Neoplasias del Timo , Adulto , Femenino , Neoplasias Cardíacas/diagnóstico por imagen , Neoplasias Cardíacas/cirugía , Hemangiosarcoma/diagnóstico , Hemangiosarcoma/cirugía , Humanos , Recurrencia Local de Neoplasia , Embarazo , Mujeres Embarazadas
2.
Cell Mol Neurobiol ; 37(5): 919-929, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27678140

RESUMEN

The CD4+CD25+ regulatory T cells (Tregs), an innate immunomodulator, suppress cerebral inflammation and maintain immune homeostasis in multiple central nervous system injury, but its role in intracerebral hemorrhage (ICH) has not been fully characterized. This study investigated the effect of Tregs on brain injury using the mouse ICH model, which is established by autologous blood infusion. The results showed that tail intravenous injection of Tregs significantly reduced brain water content and Evans blue dye extravasation of perihematoma at day (1, 3 and 7), and improved short- and long-term neurological deficits following ICH in mouse model. Tregs treatment reduced the content of pro-inflammatory cytokines interleukin (IL)-1ß, IL-6, tumor necrosis factor-α, and malondialdehyde, while increasing the superoxide dismutase (SOD) enzymatic activity at day (1, 3 and 7) following ICH. Furthermore, Tregs treatment obviously reduced the number of NF-κB+, IL-6+, TUNEL+ and active caspase-3+ cells at day 3 after ICH. These results indicate that adoptive transfer of Tregs may provide neuroprotection following ICH in mouse models.


Asunto(s)
Traslado Adoptivo , Hemorragia Cerebral/inmunología , Hemorragia Cerebral/terapia , Hematoma/inmunología , Hematoma/terapia , Inflamación/patología , Linfocitos T Reguladores/inmunología , Animales , Apoptosis , Barrera Hematoencefálica/patología , Hemorragia Cerebral/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hematoma/complicaciones , Hematoma/patología , Masculino , Malondialdehído/metabolismo , Ratones Endogámicos C57BL , Superóxido Dismutasa/metabolismo , Factor de Transcripción ReIA/metabolismo
3.
Cell Mol Neurobiol ; 37(2): 211-222, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26971524

RESUMEN

Progressive accumulation of beta-amyloid (Aß) will form the senile plaques and cause oxidative damage and neuronal cell death, which was accepted as the major pathological mechanism to the Alzheimer's disease (AD). Hence, inhibition of Aß-induced oxidative damage and neuronal cell apoptosis by agents with potential antioxidant properties represents one of the most effective strategies in combating human AD. Curcumin (Cur) a natural extraction from curcuma longa has potential of pharmacological efficacy, including the benefit to antagonize Aß-induced neurotoxicity. However, the molecular mechanism remains elusive. The present study evaluated the protective effect of Cur against Aß-induced cytotoxicity and apoptosis in PC12 cells and investigated the underlying mechanism. The results showed that Cur markedly reduced Aß-induced cytotoxicity by inhibition of mitochondria-mediated apoptosis through regulation of Bcl-2 family. The PARP cleavage, caspases activation, and ROS-mediated DNA damage induced by Aß were all significantly blocked by Cur. Moreover, regulation of p38 MAPK and AKT pathways both contributed to this protective potency. Our findings suggested that Cur could effectively suppress Aß-induced cytotoxicity and apoptosis by inhibition of ROS-mediated oxidative damage and regulation of ERK pathway, which validated its therapeutic potential in chemoprevention and chemotherapy of Aß-induced neurotoxicity.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Curcumina/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Fragmentos de Péptidos/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células PC12 , Ratas , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
4.
Neurochem Res ; 42(2): 541-551, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27868153

RESUMEN

Carnosine, an endogenous dipeptide (ß-alanyl-L-histidine), exerts multiple neuroprotective properties, but its role in intracerebral hemorrhage (ICH) remains unclear. This study investigates the effect of Carnosine on brain injury using the rat ICH model, which is established by type IV collagenase caudatum infusion. The results indicate that intraperitoneal administration of Carnosine (1000 mg/kg) significantly attenuates brain edema, blood-brain barrier (BBB) disruption, oxidative stress, microglia activation and neuronal apoptosis of perihematoma at 72 h following ICH in rats models, as convinced by preventing the disruption of tight junction protein ZO-1, occludin and claudin-5, followed by the decrease of ROS, MDA, 3-NT, 8-OHDG level and the increase of GSH-Px and SOD activity, then followed by the decline of Iba-1, ED-1, active caspase-3 and TUNEL positive cells and the decrease of IL-1ß, IL-6, TNF-α, active caspase-3 and cytochrome c level. Our results suggest that Carnosine may provide neuroprotective effect after experimental ICH in rat models.


Asunto(s)
Apoptosis/fisiología , Encéfalo/metabolismo , Carnosina/uso terapéutico , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Estrés Oxidativo/fisiología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Carnosina/farmacología , Masculino , Estrés Oxidativo/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
5.
Cell Mol Neurobiol ; 36(5): 647-55, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26224360

RESUMEN

Hypothermia treatment is one of the neuroprotective strategies that improve neurological outcomes effectively after brain damage. Minimally invasive surgery (MIS) has been an important treatment of intracerebral hemorrhage (ICH). Herein, we evaluated the neuroprotective effect and mechanism of MIS joint local cooling lavage (LCL) treatment on ICH via detecting the inflammatory responses, oxidative injury, and neuronal apoptosis around the hematoma cavity in rats. ICH model was established by type IV collagenase caudatum infusion. The rats were treated with MIS 6 h after injection, and then were lavaged by normothermic (37 °C) and hypothermic (33 °C) normal saline in brain separately. The results indicated that MIS joint LCL treatment showed enhanced therapeutic effects against ICH-induced inflammation injury and apoptosis in rats, as convinced by the decline of TUNEL-positive cells, followed by the decrease of IL-1ß and LDH and increase of IL-10 and SOD. This study demonstrated that the strategy of using MIS joint LCL may achieve enhanced neuroprotection against ICH-induced inflammation injury and apoptosis in rats with potential clinic application.


Asunto(s)
Apoptosis/efectos de los fármacos , Edema Encefálico/cirugía , Hemorragia Cerebral/complicaciones , Procedimientos Quirúrgicos Mínimamente Invasivos , Animales , Lesiones Encefálicas/cirugía , Hemorragia Cerebral/terapia , Inflamación/cirugía , Masculino , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Neuronas/metabolismo , Ratas Sprague-Dawley , Recuperación de la Función/fisiología , Irrigación Terapéutica/métodos
6.
Neurochem Res ; 41(6): 1439-47, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26846141

RESUMEN

Selenocysteine (SeC) a natural available selenoamino acid exhibits novel anticancer activities against human cancer cell lines. However, the growth inhibitory effect and mechanism of SeC in human glioma cells remain unclear. The present study reveals that SeC time- and dose-dependently inhibited U251 and U87 human glioma cells growth by induction of S-phase cell cycle arrest, followed by the marked decrease of cyclin A. SeC-induced S-phase arrest was achieved by inducing DNA damage through triggering generation of reactive oxygen species (ROS) and superoxide anion, with concomitant increase of TUNEL-positive cells and induction of p21waf1/Cip1 and p53. SeC treatment also caused the activation of p38MAPK, JNK and ERK, and inactivation of AKT. Four inhibitors of MAPKs and AKT pathways further confirmed their roles in SeC-induced S-phase arrest in human glioma cells. Our findings advance the understanding on the molecular mechanisms of SeC in human glioma management.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Daño del ADN/fisiología , Glioma/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Oncogénica v-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Selenocisteína/farmacología , Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Selenio/farmacología
7.
Cell Biol Toxicol ; 32(4): 333-45, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27184666

RESUMEN

Caudatin as one species of C-21 steroidal from Cynanchum bungei decne displays potential anticancer activity. However, the underlying mechanisms remain elusive. In the present study, the growth suppressive effect and mechanism of caudatin on human glioma U251 and U87 cells were evaluated in vitro. The results indicated that caudatin significantly inhibited U251 and U87 cell growth in both a time- and dose-dependent manner. Flow cytometry analysis revealed that caudatin-induced cell growth inhibition was achieved by induction of cell apoptosis, as convinced by the increase of Sub-G1 peak, PARP cleavage and activation of caspase-3, caspase-7 and caspase-9. Caudatin treatment also resulted in mitochondrial dysfunction which correlated with an imbalance of Bcl-2 family members. Further investigation revealed that caudatin triggered U251 cell apoptosis by inducing reactive oxygen species (ROS) generation through disturbing the redox homeostasis. Moreover, pretreatment of caspase inhibitors apparently weakens caudatin-induced cell killing, PARP cleavage and caspase activation and eventually reverses caudatin-mediated apoptosis. Importantly, caudatin significantly inhibited U251 tumour xenografts in vivo through induction of cell apoptosis involving the inhibition of cell proliferation and angiogenesis, which further validate its value in combating human glioma in vivo. Taken together, the results described above all suggest that caudatin inhibited human glioma cell growth by induction of caspase-dependent apoptosis with involvement of mitochondrial dysfunction and ROS generation.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Caspasas/metabolismo , Glioma/tratamiento farmacológico , Glicósidos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Esteroides/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Inhibidores de Caspasas/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glioma/metabolismo , Glioma/patología , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
8.
Cell Mol Neurobiol ; 35(2): 147-57, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25179154

RESUMEN

Carnosine (ß-alanyl-L-histidine) has been demonstrated to provide antioxidative and anti-apoptotic roles in the animal of ischemic brain injuries and neurodegenerative diseases. The aim of this study was to examine whether carnosine prevents subarachnoid hemorrhage (SAH)-induced early brain injury (EBI) in rats. We found that intraperitoneal administration of carnosine improved neurobehavioral deficits, attenuated brain edema and blood-brain barrier permeability, and decreased reactive oxygen species level at 48 h following SAH in rat models. Carnosine treatment increased tissue copper/zinc superoxide dismutase (CuZn-SOD) and glutathione peroxidase (GSH-Px) enzymatic activities, and reduced post-SAH elevated lactate dehydrogenase (LDH) activity, the concentration of malondialdehyde (MDA), 3-nitrotyrosine (3-NT), 8-hydroxydeoxyguanosine (8-OHDG), interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α (TNF-α) in rats. Furthermore, carnosine treatment attenuated SAH-induced microglia activation and cortical neuron apoptosis. These results indicated that administration of carnosine may provide neuroprotection in EBI following SAH in rat models.


Asunto(s)
Antioxidantes/uso terapéutico , Apoptosis , Lesiones Encefálicas/tratamiento farmacológico , Carnosina/uso terapéutico , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Antígenos Nucleares/metabolismo , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Conducta Animal , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Carnosina/farmacología , Caspasa 3/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Citocinas/metabolismo , ADN/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Glutatión Peroxidasa/metabolismo , Etiquetado Corte-Fin in Situ , Lípidos/química , Masculino , Microglía/efectos de los fármacos , Microglía/patología , Proteínas del Tejido Nervioso/metabolismo , Oxidación-Reducción/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/patología , Superóxido Dismutasa/metabolismo
9.
Cell Mol Neurobiol ; 35(7): 953-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25860846

RESUMEN

Caudatin, one of the species of C-21 steroidal glycosides mainly isolated from the root of Cynanchum bungei Decne, exhibits potent anticancer activities. However, the mechanism remains poorly defined. In the present study, the growth inhibitory effect and mechanism of caudatin on human glioma cells were evaluated in vitro. The results revealed that caudatin time- and dose-dependently inhibited U251 and U87 cells growth. Flow cytometry analysis indicated that caudatin-induced growth inhibition against U251 and U87 cells was mainly achieved by the induction of G0/G1 and S-phase cell cycle arrest through triggering DNA damage, as convinced by the up-regulation of p53, p21, and histone phosphorylation, as well as the down-regulation of cyclin D1. Moreover, caudatin treatment also triggered the activation of ERK and inactivation of AKT pathway. LY294002 (an AKT inhibitor) addition enhanced caudation-induced AKT inhibition, indicating that caudatin inhibited U251 cells growth in an AKT-dependent manner. Taken together, these results indicate that caudatin may act as a novel cytostatic reagent against human glioma cells through the induction of DNA damage-mediated cell cycle arrest with the involvement of modulating MAPK and AKT pathways.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Glioma/tratamiento farmacológico , Glicósidos/uso terapéutico , Inhibidores de Crecimiento/uso terapéutico , Esteroides/uso terapéutico , Puntos de Control del Ciclo Celular/fisiología , Línea Celular Tumoral , Daño del ADN/fisiología , Relación Dosis-Respuesta a Droga , Glioma/metabolismo , Glicósidos/química , Glicósidos/farmacología , Inhibidores de Crecimiento/farmacología , Humanos , Esteroides/química , Esteroides/farmacología
10.
Cell Mol Neurobiol ; 35(4): 543-53, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25527033

RESUMEN

Oxidative stress plays an important role in the pathogenesis of early brain injury (EBI) following subarachnoid hemorrhage (SAH). The aim of this study was to assess whether cysteamine prevents post-SAH oxidative stress injury via its antioxidative and anti-apoptotic effects. It was observed that intraperitoneal administration of cysteamine (20 mg/kg/day) could significantly alleviate EBI (including neurobehavioral deficits, brain edema, blood-brain barrier permeability, and cortical neuron apoptosis) after SAH in rats. Meanwhile, cysteamine treatment reduced post-SAH elevated the reactive oxygen species level, the concentration of malondialdehyde, 3-nitrotyrosine, and 8-hydroxydeoxyguanosine and increased the glutathione peroxidase enzymatic activity, the concentration of glutathione and brain-derived neurotrophic factor in brain cortex at 48 h after SAH. These results indicated that administration of cysteamine may ameliorate EBI and provide neuroprotection after SAH in rat models.


Asunto(s)
Apoptosis , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Cisteamina/uso terapéutico , Estrés Oxidativo , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/patología , Animales , Apoptosis/efectos de los fármacos , Conducta Animal , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Caspasa 3/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Cisteamina/farmacología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Etiquetado Corte-Fin in Situ , Masculino , Estrés Oxidativo/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Hemorragia Subaracnoidea/complicaciones
11.
Cell Mol Neurobiol ; 35(7): 995-1001, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25895624

RESUMEN

Cisplatin-based chemotherapy in clinic is severely limited by its adverse effect, including neurotoxicity. Oxidative damage contributes to cisplatin-induced neurotoxicity, but the mechanism remains unclearly. Cyanidin, a natural flavonoid compound, exhibits powerful antioxidant activity. Hence, we investigated the protective effects of cyanidin on PC12 cells against cisplatin-induced neurotoxicity and explored the underlying mechanisms. The results showed that cisplatin-induced cytotoxicity was completely reversed by cyanidin through inhibition of PC12 cell apoptosis, as proved by the attenuation of Sub-G1 peak, PARP cleavage, and caspases-3 activation. Mechanistically, cyanidin significantly inhibited reactive oxygen species (ROS)-induced DNA damage in cisplatin-treated PC12 cells. Our findings revealed that cyanidin as an apoptotic inhibitor effectively blocked cisplatin-induced neurotoxicity through inhibition of ROS-mediated DNA damage and apoptosis, predicating its therapeutic potential in prevention of chemotherapy-induced neurotoxicity. Cisplatin caused DNA damage, activated p53, and subsequently induced PC12 cells apoptosis by triggering ROS overproduction. However, cyanidin administration effectively inhibited DNA damage, attenuated p53 phosphorylation, and eventually reversed cisplatin-induced PC12 cell apoptosis through inhibition ROS accumulation.


Asunto(s)
Antocianinas/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Cisplatino/toxicidad , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Células PC12 , Ratas
12.
Neurochem Res ; 40(6): 1121-32, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25846008

RESUMEN

Activation of metabotropic glutamate receptor 5 (mGluR5) provided neuroprotection in multiple central nervous system injury, but the roles of mGluR5 in subarachnoid hemorrhage (SAH) remain unclear. In present study, we aimed to evaluate whether activation of mGluR5 attenuates early brain injury (EBI) after experimental SAH in rats. We found that selective mGluR5 orthosteric agonist CHPG or positive allosteric modulator VU0360172 administration significantly improves neurological function and attenuates brain edema at 24 h after SAH. Furthermore, mGluR5 obviously expresses in activated microglia (ED-1 positive) after SAH. CHPG or VU0360172 administration significantly reduces the numbers of activated microglia and the protein and mRNA levels of pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α at 24 h after SAH. Moreover, CHPG or VU0360172 administration obviously reduces the number of TUNEL-positive cells and active caspase-3/NeuN-positive neurons in cortex at 24 h after SAH. CHPG or VU0360172 administration significantly up-regulates the expression of Bcl-2, and down-regulates the expression of Bax and active caspase-3, which in turn increases the ratio of Bcl-2/Bax. Our results indicate that activation of mGluR5 attenuates microglial activation and neuronal apoptosis, and improves neurological function in EBI after SAH.


Asunto(s)
Apoptosis/efectos de los fármacos , Microglía/patología , Neuronas/patología , Receptor del Glutamato Metabotropico 5/metabolismo , Hemorragia Subaracnoidea/patología , Animales , Conducta Animal/efectos de los fármacos , Edema Encefálico/prevención & control , Caspasa 3/biosíntesis , Caspasa 3/genética , Corteza Cerebral/patología , Ciclina D1/biosíntesis , Ciclina D1/genética , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Agonistas de Aminoácidos Excitadores/uso terapéutico , Glicina/análogos & derivados , Glicina/uso terapéutico , Activación de Macrófagos/efectos de los fármacos , Masculino , Fenilacetatos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/mortalidad , Hemorragia Subaracnoidea/psicología , Proteína X Asociada a bcl-2/biosíntesis , Proteína X Asociada a bcl-2/genética
15.
ACS Omega ; 8(22): 19692-19704, 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37305289

RESUMEN

Extracellular signal-regulated kinase 1 and 2 (Erk1/2) signaling has been shown to be involved in brain injury after subarachnoid hemorrhage (SAH). A first-in-human phase I study reported that ravoxertinib hydrochloride (RAH), a novel Erk1/2 inhibitor, has an acceptable safety profile and pharmacodynamic effects. Here, we showed that the level of Erk1/2 phosphorylation (p-Erk1/2) was significantly increased in the cerebrospinal fluid (CSF) of aneurysmal subarachnoid hemorrhage (aSAH) patients who developed poor outcomes. In a rat SAH model that was produced by the intracranial endovascular perforation method, western blot observed that the level of p-Erk1/2 was also increased in the CSF and basal cortex, showing a similar trend with aSAH patients. Immunofluorescence and western blot indicated that RAH treatment (i.c.v injection, 30 min post-SAH) attenuates the SAH-induced increase of p-Erk1/2 at 24 h in rats. RAH treatment can improve experimental SAH-induced long-term sensorimotor and spatial learning deficits that are evaluated by the Morris water maze, rotarod test, foot-fault test, and forelimb placing test. Moreover, RAH treatment attenuates neurobehavioral deficits, the blood-brain barrier damage, and cerebral edema at 72 h after SAH in rats. Furthermore, RAH treatment decreases the SAH-elevated apoptosis-related factor active caspase-3 and the necroptosis-related factor RIPK1 expression at 72 h in rats. Immunofluorescence analysis showed that RAH attenuated neuronal apoptosis but not neuronal necroptosis in the basal cortex at 72 h after SAH in rats. Altogether, our results suggest that RAH improves long-term neurologic deficits through early inhibition of Erk1/2 in experimental SAH.

16.
Front Cell Neurosci ; 16: 878673, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35573833

RESUMEN

Cell apoptosis plays an important role in early brain injury (EBI) after subarachnoid hemorrhage (SAH). Heat shock protein 27 (HSP27), a member of the small heat shock protein (HSP) family, is induced by various stress factors and exerts protective role on cells. However, the role of HSP27 in brain injury after SAH needs to be further clarified. Here, we reported that HSP27 level of cerebrospinal fluid (CSF) is increased obviously at day 1 in patients with aneurysmal SAH (aSAH) and related to the grades of Hunt and Hess (HH), World Federation of Neurological Surgeons (WFNS), and Fisher score. In rat SAH model, HSP27 of CSF is first increased and then obviously declined; overexpression of HSP27, not knockdown of HSP27, attenuates SAH-induced neurological deficit and cell apoptosis in the basal cortex; and overexpression of HSP27 effectively suppresses SAH-elevated activation of mitogen-activated protein Kinase Kinase 4 (MKK4), the c-Jun N-terminal kinase (JNK), c-Jun, and caspase-3. In an in vitro hemolysate-damaged cortical neuron model, HSP2765-90 peptide effectively inhibits hemolysate-induced neuron death. Furthermore, TAT-HSP2765-90 peptide, a fusion peptide consisting of trans-activating regulatory protein (TAT) of HIV and HSP2765-90 peptide, effectively attenuates SAH-induced neurological deficit and cell apoptosis in the basal cortex of rats. Altogether, our results suggest that TAT-HSP27 peptide improves neurologic deficits via reducing apoptosis.

17.
Biochim Biophys Acta ; 1804(4): 929-40, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20079886

RESUMEN

Enhanced salt tolerance of rice seedlings by abscisic acid (ABA) pretreatment was observed from phenotypic and physiological analyses. Total proteins from rice roots treated with ABA plus subsequent salt stress were analyzed by using proteomics method. Results showed that, 40 protein spots were uniquely upregulated in the seedlings under the condition of ABA pretreatment plus subsequent salt stress, whereas only 16 under the condition of salt treatment. About 78% (31 spots) of the 40 protein spots were only upregulated in the presence of the subsequent salt stress, indicating that plants might have an economical strategy to prevent energy loss under a false alarm. The results also showed that more enzymes involved in energy metabolism, defense, primary metabolism, etc. were upregulated uniquely in ABA-pretreated rice seedlings, suggesting more abundant energy supply, more active anabolism (nitrogen, nucleotide acid, carbohydrate, etc), and more comprehensive defense systems in ABA-pretreated seedlings than in salt stressed ones.


Asunto(s)
Ácido Abscísico/farmacología , Oryza/efectos de los fármacos , Oryza/metabolismo , Reguladores del Crecimiento de las Plantas/farmacología , Proteínas de Plantas/metabolismo , Proteoma/metabolismo , Tolerancia a la Sal/efectos de los fármacos , Ácido Abscísico/metabolismo , Metabolismo Energético/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Oryza/crecimiento & desarrollo , Fenotipo , Reguladores del Crecimiento de las Plantas/metabolismo , Raíces de Plantas/efectos de los fármacos , Raíces de Plantas/metabolismo , Proteómica , Plantones/efectos de los fármacos , Plantones/crecimiento & desarrollo , Plantones/metabolismo , Estrés Fisiológico , Regulación hacia Arriba/efectos de los fármacos
18.
Acta Neurochir Suppl ; 110(Pt 2): 49-53, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21125445

RESUMEN

Substances and fluid in the brain and subarachnoid spaces may be drained into extracranial lymphatics. This study aimed to investigate the possible role of cerebral lymphatic drainage in the process of cerebral injury following subarachnoid hemorrhage (SAH). Wistar rats were divided into non-SAH, SAH, and SAH plus cervical lymphatic blockage (SAH + CLB) groups. Autologous arterial hemolysate was injected into rats' cisterna magna to induce SAH. At time of 24 and 72 h after SAH, the rats were sacrificed for serum lactate dehydrogenase (LDH) activity, brain tissue superoxide dismutase (SOD) activity, and brain tissue malonaldehyde (MDA) content detection. It was found that serum LDH activity increased in rats of SAH group comparing with non-SAH group. SAH also resulted in decreased brain tissue SOD activity and increased brain tissue MDA content. In rats of SAH + CLB group, the increase of serum LDH activity was to a lager extent. Meanwhile, brain tissue SOD activity decreased and MDA content increased to a lager extent, as compared with SAH group. It was concluded that blockage of cerebral lymphatic drainage deteriorates cerebral oxidative injury after SAH, indicating cerebral lymphatic drainage may exert intrinsic protective effects against cerebral injury following SAH.


Asunto(s)
Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Corteza Cerebral/patología , Vasos Linfáticos/lesiones , Hemorragia Subaracnoidea/complicaciones , Superóxido Dismutasa/metabolismo , Animales , Análisis de los Gases de la Sangre/métodos , Presión Sanguínea/fisiología , Lesiones Encefálicas/sangre , Modelos Animales de Enfermedad , Femenino , L-Lactato Deshidrogenasa/sangre , Vasos Linfáticos/patología , Masculino , Malondialdehído/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
19.
Exp Ther Med ; 22(6): 1364, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34659510

RESUMEN

Chemoresistance is the primary cause of the poor outcome of glioblastoma multiforme (GBM) therapy. Leucine-rich repeat-containing G-protein coupled receptor 6 (LGR6) is involved in the growth and proliferation of several types of cancer, including gastric cancer and ovarian cancer. Therefore, the aim of the present study was to investigate the role of LGR6 in GBM malignancy and chemoresistance. Cell counting kit-8 and Matrigel®-Transwell assays were conducted to assess GBM cell viability and invasion. The effect of LGR6 on cell cycle progression and activation of Akt signaling was analyzed by performing propidium iodide staining and western blotting, respectively. The results demonstrated that LGR6, a microRNA-1236-3p target candidate, promoted GBM cell viability and invasion, and mediated temozolomide sensitivity in SHG-44 and U251 GBM cells. In addition, LGR6 triggered the activation of the Akt signaling pathway during GBM progression. Collectively, the results of the present study suggested that LGR6 promoted GBM malignancy and chemoresistance, at least in part, by activating the Akt signaling pathway. The results may aid with the identification of a novel therapeutic target and strategy for GBM.

20.
Transl Stroke Res ; 12(6): 1067-1080, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33713028

RESUMEN

Excessive glutamate leading to excitotoxicity worsens brain damage after SAH and contributes to long-term neurological deficits. The drug ifenprodil is a non-competitive antagonist of GluN1-GluN2B N-methyl-d-aspartate (NMDA) receptor, which mediates excitotoxic damage in vitro and in vivo. Here, we show that cerebrospinal fluid (CSF) glutamate level within 48 h was significantly elevated in aSAH patients who later developed poor outcome. In rat SAH model, ifenprodil can improve long-term sensorimotor and spatial learning deficits. Ifenprodil attenuates experimental SAH-induced neuronal death of basal cortex and hippocampal CA1 area, cellular and mitochondrial Ca2+ overload of basal cortex, blood-brain barrier (BBB) damage, and cerebral edema of early brain injury. Using in vitro models, ifenprodil declines the high-concentration glutamate-mediated intracellular Ca2+ increase and cell apoptosis in primary cortical neurons, reduces the high-concentration glutamate-elevated endothelial permeability in human brain microvascular endothelial cell (HBMEC). Altogether, our results suggest ifenprodil improves long-term neurologic deficits through antagonizing glutamate-induced excitotoxicity.


Asunto(s)
Ácido Glutámico , Hemorragia Subaracnoidea , Animales , Barrera Hematoencefálica/metabolismo , Ácido Glutámico/toxicidad , Humanos , Piperidinas/farmacología , Piperidinas/uso terapéutico , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA