Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Sci Data ; 11(1): 449, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702307

RESUMEN

In the context of China's freshwater crisis high-resolution data are critical for sustainable water management and economic growth. Yet there is a dearth of data on water withdrawal and scarcity regardless of whether total or subsector amount, for prefectural cities. In administrative and territorial scope, we accounted for water withdrawal of all 63 economic-socio-environmental sectors for all 343 prefectural cities in China, based on a general framework and 2015 data. Spatial and economic-sector resolution is improved compared with previous studies by partitioning general sectors into industrial and agricultural sub-sectors. Construction of these datasets was based on selection of 16 driving forces. We connected a size indicator with corresponding water-withdrawal efficiency. We further accounted for total blue-water withdrawal and quantitative water scarcity status. Then we compared different scopes and methods of official accounts and statistics from various water datasets. These disaggregated and complete data could be used in input-output models for municipal design and governmental planning to help gain in-depth insights into subsector water-saving priorities from local economic activities.

2.
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.

4.
Front Mol Neurosci ; 15: 971361, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36046710

RESUMEN

Lysosomal-associated transmembrane protein 5 (LAPTM5) has been demonstrated to be involved in regulating immunity, inflammation, cell death, and autophagy in the pathophysiological processes of many diseases. However, the function of LAPTM5 in cerebral ischemia-reperfusion (I/R) injury has not yet been reported. In this study, we found that LAPTM5 expression was dramatically decreased during cerebral I/R injury both in vivo and in vitro. LAPTM5 knockout (KO) mice were compared with a control, and they showed a larger infarct size and more serious neurological dysfunction after transient middle cerebral artery occlusion (tMCAO) treatment. In addition, inflammatory response and apoptosis were exacerbated in these processes. Furthermore, gain- and loss-of-function investigations in an in vitro model revealed that neuronal inflammation and apoptosis were aggravated by LAPTM5 knockdown but mitigated by its overexpression. Mechanistically, combined RNA sequencing and experimental verification showed that the apoptosis signal-regulating kinase 1 (ASK1)-c-Jun N-terminal kinase (JNK)/p38 pathway was mainly involved in the detrimental effects of LAPTM5 deficiency following I/R injury. Specifically, LAPTM5 directly interacts with ASK1, leading to decreased ASK1 N-terminal dimerization and the subsequent reduced activation of downstream JNK/p38 signaling. In conclusion, LAPTM5 was demonstrated to be a novel modulator in the pathophysiology of brain I/R injury, and targeting LAPTM5 may be feasible as a stroke treatment.

5.
Neurosurg Rev ; 45(6): 3717-3728, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36169785

RESUMEN

Recent studies have demonstrated that hyperglycemia may result in a poor prognosis following aneurysmal subarachnoid hemorrhage (aSAH). However, the association between hyperglycemia and the clinical outcome of aSAH has not been clearly established thus far. Therefore, we performed a systematic review and meta-analysis to investigate the association between hyperglycemia and the development of aSAH. We completed a literature search in four databases (PubMed, EMBASE, Cochrane Library, and Web of Science) up to November 1, 2021, including all eligible studies investigating the prognostic value of hyperglycemia in patients with aSAH. We performed a quality assessment of included studies using the Newcastle-Ottawa Quality Assessment Scale. The pooled odds ratios (ORs) with corresponding 95% confidence intervals (CIs) were calculated to assess the association of hyperglycemia in aneurysmal subarachnoid hemorrhage. A total of 35 studies with 11,519 patients were finally included in the meta-analysis. Nineteen studies reported the association between hyperglycemia and poor outcome, 12 studies reported the association between hyperglycemia and all-cause mortality, 7 studies reported the association between hyperglycemia and cerebral vasospasm, and 9 studies reported the association between hyperglycemia and cerebral infarction. The pooled data of these studies suggested that hyperglycemia was significantly associated with poor functional outcomes (odds ratio [OR], 1.29; 95% confidence interval [CI], 1.17-1.42; P < 0.00001; I2 = 83%), all-cause mortality (OR, 1.02; 95% CI, 1.01-1.04; P = 0.0006; I2 = 89%), cerebral vasospasm (OR, 1.02; 95% CI, 1.01-1.02; P = 0.0002; I2 = 35%), and cerebral infarction (OR, 1.16; 95% CI, 1.09-1.23; P < 0.00001; I2 = 10%) in aSAH patients. These findings suggested that assessing for hyperglycemia at admission may help clinicians to identify critically ill patients and complete patient stratification early, which may achieve better management and improve the prognosis of patients with aSAH.


Asunto(s)
Hiperglucemia , Hemorragia Subaracnoidea , Vasoespasmo Intracraneal , Humanos , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/diagnóstico , Hemorragia Subaracnoidea/cirugía , Vasoespasmo Intracraneal/etiología , Pronóstico , Infarto Cerebral/complicaciones , Hiperglucemia/complicaciones
6.
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.

7.
Front Pharmacol ; 13: 848529, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35529450

RESUMEN

Edaravone dexborneol is a novel neuroprotective drug that comprises edaravone and (+)-borneol in a 4:1 ratio. Phase II and III studies have demonstrated that Chinese patients treated with edaravone dexborneol within 48 h of AIS onset have better functional outcomes than those treated with edaravone alone. However, the effect of edaravone dexborneol on subarachnoid hemorrhage (SAH) has not yet been elucidated. This study aimed to investigate the therapeutic effects of edaravone dexborneol on SAH-induced brain injury and long-term behavioral deficits and to explore the possible mechanisms. The experimental rat SAH model was induced by an intraluminal puncture of the left middle cerebral artery (MCA). Edaravone dexborneol or edaravone at a clinical dose was infused into the tail vein for 3 days post-SAH surgery. Behavioral outcomes were assessed by a modified Garcia scoring system and rotarod, foot-fault, and corner tests. Immunofluorescence, Western blot, and ELISA methods were used to evaluate neuronal damage and oxidative stress. Our results showed that a post-SAH therapeutic regimen with edaravone dexborneol helped improve neurological function up to 21 days after SAH surgery and demonstrated a greater beneficial effect than edaravone alone, accompanied by an obvious inhibition of neuronal apoptosis in the CA1 hippocampus and basal cortex regions. Mechanistically, edaravone dexborneol not only suppressed the lipid peroxidation product malondialdehyde (MDA) but also improved the total antioxidant capability (TAC) 3 days after SAH. Notably, edaravone dexborneol treatment significantly inhibited the expression of another lipid peroxidation product, 4-hydroxynonenal (4-HNE), in the CA1 hippocampus and basal cortex, which are vital participants in the process of neuronal oxidative damage and death after SAH because of their acute cytotoxicity. Together, our results demonstrate that edaravone dexborneol confers neuroprotection and stabilizes long-term behavioral ability after SAH injury, possibly by suppressing 4-HNE-associated oxidative stress. These results may help develop new clinical strategies for SAH treatment.

8.
J Neurochem ; 160(3): 392-411, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34837397

RESUMEN

TBC1Domain Family Member 25 (TBC1D25) is a protein that contains a TBC/RAB-GTPase activating protein (GAP) domain, which was shown to participate in autophagy in previous studies. However, the role of TBC1D25 in cerebral ischemia-reperfusion (I/R) injury remains unknown. In this study, we found that the mRNA and protein expression levels of TBC1D25 decreased in mouse brain after I/R injury and primary cortical neurons treated with oxygen and glucose deprivation/reoxygenation (OGD/R). Then TBC1D25 knockout (KO) mice were applied to demonstrate that TBC1D25 ablation aggravated cerebral I/R-induced neuronal loss and infarct size. In addition, neuronal apoptosis and inflammation were significantly potentiated in the TBC1D25-KO group. In in vitro OGD/R model, TBC1D25 knockdown can attenuate neuronal cell viability and aggravate the process of inflammation and apoptosis. Conversely, over-expression of TBC1D25 in primary neurons ameliorated the aforementioned processes. Mechanistically, RNA-sequencing (RNA-seq) analysis revealed mitogen-activated protein kinase (MAPK) signaling pathway was the most significant pathway that contributed to TBC1D25-mediated brain I/R injury process. Through experimental verification, TBC1D25 deficiency increased the phosphorylation of the transforming growth factor-ß-activated kinase 1 (TAK1)-c-Jun N-terminal kinase (JNK)/p38 axis in neurons during the brain I/R injury. Furthermore, we found that TAK1 blockade abrogated the apoptosis and inflammatory response produced by TBC1D25 knockdown in vitro. In conclusion, this study is the first to demonstrate the functional significance of TBC1D25 in the pathophysiology of brain I/R injury, and the protective mechanism of TBC1D25 is dependent on the TAK1-JNK/p38 pathway.


Asunto(s)
Isquemia Encefálica/genética , Proteínas Activadoras de GTPasa/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Quinasas Quinasa Quinasa PAM/genética , Daño por Reperfusión/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Animales , Apoptosis , Isquemia Encefálica/fisiopatología , Proteínas Activadoras de GTPasa/deficiencia , Glucosa/deficiencia , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/fisiopatología , Inflamación/genética , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , RNA-Seq , Daño por Reperfusión/fisiopatología
9.
Molecules ; 26(9)2021 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-34064401

RESUMEN

Hydrogel adhesives are attractive for applications in intelligent soft materials and tissue engineering, but conventional hydrogels usually have poor adhesion. In this study, we designed a strategy to synthesize a novel adhesive with a thin hydrogel adhesive layer integrated on a tough substrate hydrogel. The adhesive layer with positive charges of ammonium groups on the polymer backbones strongly bonds to a wide range of nonporous materials' surfaces. The substrate layer with a dual hydrogen bond system consists of (i) weak hydrogen bonds between N,N-dimethyl acrylamide (DMAA) and acrylic acid (AAc) units and (ii) strong multiple hydrogen bonds between 2-ureido-4[1H]-pyrimidinone (UPy) units. The dual hydrogen-bond network endowed the hydrogel adhesives with unique mechanical properties, e.g., toughness, highly stretchability, and insensitivity to notches. The hydrogel adhesion to four types of materials like glass, 316L stainless steel, aluminum, Al2O3 ceramic, and two biological tissues including pig skin and pig kidney was investigated. The hydrogel bonds strongly to dry solid surfaces and wet tissue, which is promising for biomedical applications.


Asunto(s)
Hidrogeles/química , Acrilamidas/química , Acrilatos/química , Adhesividad , Animales , Enlace de Hidrógeno , Porcinos
10.
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
11.
Carbohydr Res ; 501: 108275, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33657498

RESUMEN

In order to achieve efficient delivery of methotrexate (MTX), thymine-chitosan nanoparticles (Thy-Cs NPs) were prepared, and further decorated with lactobionic acid (LA) to obtain tumor-targeting nanoparticles (LA-Thy-Cs NPs). These nanoparticles possessed a regular spherical structure with the average size about 190-250 nm and narrow size distribution, which were kinetically stable in the physiological environment. Due to electrostatic interactions and multiple hydrogen-bonding interactions between MTX and carriers, MTX was loaded into Thy-Cs NPs with high drug loading content (~20%). MTX release from Thy-Cs NPs was significantly accelerated in the mildly acidic environment due to the destruction of two types of non-covalent interactions. In vitro cell experiments demonstrated that LA-Thy-Cs NPs could be efficiently internalized into hepatoma carcinoma cells, leading to higher cytotoxicity. Moreover, MTX-loaded LA-Thy-Cs NPs performed an enhanced growth inhibition in three-dimensional multicellular tumor spheroids. Thus, the LA decorated thymine-chitosan nanocarriers can be a promising candidate for efficient delivery of MTX.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Quitosano/química , Disacáridos/química , Sistemas de Liberación de Medicamentos , Metotrexato/farmacología , Nanopartículas/química , Timina/química , Antimetabolitos Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos/química , Ensayos de Selección de Medicamentos Antitumorales , Células Hep G2 , Humanos , Metotrexato/química , Conformación Molecular
12.
CNS Neurosci Ther ; 27(1): 82-91, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33280237

RESUMEN

The aim of present study was to explore whether 2-cyano-3, 12-dioxooleana-1, 9-dien-28-oic acid (CDDO)-ethylamide (CDDO-EA) attenuates cerebral ischemic injury and its possible mechanisms using a middle cerebral artery occlusion (MCAO) model in C57BL/6 mice. Our results showed that intraperitoneal injection (i.p.) of CDDO-EA (2 and 4 mg/kg) augmented NFE2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression in ischemic cortex after MCAO. Moreover, CDDO-EA (2 mg/kg, i.p.) significantly enhanced Nrf2 nuclear accumulation, associated with increased cytosolic HO-1 expression, reduced neurological deficit and infarct volume as well as neural apoptosis, and shifted polarization of microglia/macrophages toward an antiinflammatory M2 phenotype in ischemic cortex after MCAO. Using an in vitro model, we confirmed that CDDO-EA (100 µg/mL) increased HO-1 expression and primed microglial polarization toward M2 phenotype under inflammatory stimulation in BV2 microglial cells. These findings suggest that a novel Nrf2 activator CDDO-EA confers neuroprotection against ischemic injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Ácido Oleanólico/análogos & derivados , Animales , Células Cultivadas , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Ácido Oleanólico/farmacología , Ácido Oleanólico/uso terapéutico , Fenotipo
13.
Water Res ; 184: 116163, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32758721

RESUMEN

The water planetary boundary (PB) has attracted wide academic attention, but empirical water footprint research that accommodates local biophysical boundaries remains scarce. Here we develop two novel quantitative footprint indicators, the water exceedance footprint and the surplus water footprint. The first measures the amount of excessive water withdrawal (exceeded amount of water withdrawn against local water PBs) and the latter evaluates the potential of surplus water that can be sustainably utilised (amount of surplus water available within local water PBs). We quantify the extent to which demand for goods and services in Chinese provinces and cities are driving excessive withdrawal of local and global water resources. We investigate both territorial and consumption-based water withdrawal deficit and surplus against local water withdrawal PBs. We also trace how PB-exceeded water and surplus water are appropriated for producing certain commodities. In 2015, China's domestic water exceedance reaches 101 km3 while the total water exceedance footprint is 92 km3. We find that 47% of domestic excessive water withdrawal is associated with interprovincial trade. Exceeded water transfers were dominated by agricultural trade from the drier North to the wetter South. A revised virtual water trade network informed by exceedance and surplus water footprint metrics could help address sustainability concerns that arise from the trade of water-intensive commodities. Our findings highlight that policy targets need to accommodate PB exceedance of both direct and virtual water use.


Asunto(s)
Abastecimiento de Agua , Agua , China , Ciudades , Recursos Hídricos
14.
ACS Chem Neurosci ; 11(18): 2869-2880, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32786302

RESUMEN

Aneurysmal subarachnoid hemorrhage (SAH) causes permanent neurological sequelae, but the underlying mechanism needs to be further clarified. Here, we show that inhibition of metabotropic glutamate receptor 1 (mGluR1) with negative allosteric modulator JNJ16259685 improves long-term neurobehavioral outcomes in an endovascular perforation model of SAH. JNJ16259685 improves cerebrovascular dysfunction through attenuation of cerebral blood flow (CBF) reduction, cerebral vasoconstrictio, and microthrombosis formation in a rat SAH model. Moreover, JNJ16259685 reduces experimental SAH-induced long-term neuronal damage through alleviation of neuronal death and degeneration. Mechanically, JNJ16259685 maintains phosphorylation of endothelial NO synthase (eNOS) and vasodilator-stimulated phosphoprotein (VASP) and decreases apoptosis-related factors Bax, active caspase-9, and active caspase-3 following experimental SAH. Altogether, our results suggest JNJ16259685 improves long-term functional impairment through neurovascular protection.


Asunto(s)
Receptores de Glutamato Metabotrópico , Hemorragia Subaracnoidea , Vasoespasmo Intracraneal , Animales , Modelos Animales de Enfermedad , Ratas , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/tratamiento farmacológico
15.
ACS Omega ; 5(25): 15385-15389, 2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32637812

RESUMEN

Delayed cerebral ischemia (DCI) is an important complication after aneurysmal subarachnoid hemorrhage (aSAH). Early identification of cerebrospinal fluid (CSF) markers is helpful for warning of impending DCI. This study assessed whether early high CSF glutamate levels can be observed in aSAH patients who later developed DCI. In this prospective clinical study, patients with normal pressure hydrocephalus or aSAH were enrolled. We found that the early CSF levels of glutamate were significantly elevated in aSAH patients compared to patients with normal pressure hydrocephalus. There was a significant difference in early CSF levels of glutamate between aSAH patients without DCI and with DCI. The early CSF levels of glutamate are significantly related to the Hunt and Hess grade, the World Federation of Neurological Surgeons (WFNS) grade, and the modified Fisher score on admission and occurrence of DCI in aSAH patients. Preliminary evidence of this study suggests that early high CSF glutamate levels are correlated with DCI in aSAH patients.

16.
World Neurosurg ; 139: e508-e516, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32311566

RESUMEN

BACKGROUND: Intracranial primary central nervous system lymphoma (PCNSL) is a rare aggressive malignant tumor with poor prognosis. The effect of surgical resection on intracranial PCNSL is still controversial. This study investigates the efficacy and safety of surgical resection, as well as to analyze the clinical characteristics and prognostic factors of intracranial PCNSL. METHODS: The clinical materials of 89 consecutive patients with intracranial PCNSL were analyzed retrospectively. Outcome in survival was assessed by progression-free survival (PFS) and overall survival (OS). Univariate and multivariate analyses were performed for various potential prognostic factors to identify independent prognostic factors of intracranial PCNSL. RESULTS: Among the 89 patients, gross total resection (GTR) was achieved in 57 patients (64.0%), subtotal resection (STR) in 14 patients (15.8%), and biopsy in 18 patients (20.2%).The PFS and OS at 2 years were estimated at 32.3% and 74.1%, respectively. The median PFS was 20 months (95% confidence interval, 16-23) and the median OS was 32 months (95% confidence interval, 25-38). Patients with surgical resection (GTR and STR) had better PFS than those with biopsy, and the difference of PFS was statistically significant (P = 0.007). However, the difference of OS was not statistically significant (P = 0.062). Multivariate analysis showed that invasion of deep structure was the only independent risk factor for intracranial PCNSL. Eleven patients (12.4%) had surgical complications, mainly including limb weakness and visual field defect. CONCLUSIONS: For intracranial PCNSL, surgical excision can improve PFS but not OS. Invasion of deep structure was the only independent risk factor for intracranial PCNSL.


Asunto(s)
Neoplasias del Sistema Nervioso Central/cirugía , Linfoma/cirugía , Procedimientos Neuroquirúrgicos/métodos , Adolescente , Adulto , Anciano , Neoplasias del Sistema Nervioso Central/diagnóstico por imagen , Quimioradioterapia , Niño , Preescolar , Terapia Combinada , Femenino , Humanos , Lactante , Estado de Ejecución de Karnofsky , Linfoma/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Procedimientos Neuroquirúrgicos/efectos adversos , Seguridad del Paciente , Pronóstico , Supervivencia sin Progresión , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento , Adulto Joven
17.
Transl Stroke Res ; 11(4): 799-811, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31833035

RESUMEN

The blood-brain barrier (BBB) disruption leads to the vasogenic brain edema and contributes to the early brain injury (EBI) after subarachnoid hemorrhage (SAH). However, the mechanisms underlying the BBB damage following SAH are poorly understood. Here we reported that the neurotransmitter glutamate of cerebrospinal fluid (CSF) was dramatically increased in SAH patients with symptoms of cerebral edema. Using the rat SAH model, we found that SAH caused the increase of CSF glutamate level and BBB permeability in EBI, intracerebroventricular injection of exogenous glutamate deteriorated BBB damage and cerebral edema, while intraperitoneally injection of metabotropic glutamate receptor 1(mGluR1) negative allosteric modulator JNJ16259685 significantly attenuated SAH-induced BBB damage and cerebral edema. In an in vitro BBB model, we showed that glutamate increased monolayer permeability of human brain microvascular endothelial cells (HBMEC), whereas JNJ16259685 preserved glutamate-damaged BBB integrity in HBMEC. Mechanically, glutamate downregulated the level and phosphorylation of vasodilator-stimulated phosphoprotein (VASP), decreased the tight junction protein occludin, and increased AQP4 expression at 72 h after SAH. However, JNJ16259685 significantly increased VASP, p-VASP, and occludin, and reduced AQP level at 72 h after SAH. Altogether, our results suggest an important role of glutamate in disruption of BBB function and inhibition of mGluR1 with JNJ16259685 reduced BBB damage and cerebral edema after SAH.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Edema Encefálico/metabolismo , Quinolinas/administración & dosificación , Receptores de Glutamato Metabotrópico/agonistas , Hemorragia Subaracnoidea/complicaciones , Animales , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/líquido cefalorraquídeo , Edema Encefálico/etiología , Permeabilidad Capilar/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Ácido Glutámico/líquido cefalorraquídeo , Humanos , Masculino , Persona de Mediana Edad , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/líquido cefalorraquídeo
18.
ACS Chem Neurosci ; 10(1): 746-756, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30339347

RESUMEN

Excessive glutamate-mediated overactivation of metabotropic glutamate receptor 1 (mGluR1) plays a leading role in neuronal apoptosis following subarachnoid hemorrhage (SAH). TAT-mGluR1, a fusion peptide consisting of a peptide spanning the calpain cleavage site of mGluR1α and the trans-activating regulatory protein (TAT) of HIV, effectively blocks mGluR1α truncation and protects neurons against excitotoxic damage. This study investigated the effects of TAT-mGluR1 on neuronal apoptosis in the rat SAH model. Here, we report that SAH caused activation of calpain and truncation of mGluR1α; intraperitoneally administered TAT-mGluR1 did not affect calpain activity, while it blocked truncation of mGluR1α after SAH. Intraperitoneally administered FITC-labeled TAT-mGluR1 was colocalized with mGluR1α in thecortex after SAH. Furthermore, TAT-mGluR1 significantly improved the neurological deficit, increased p-PI3K, p-Akt, and p-GSK3ß, downregulated Bax, upregulated Bcl-2, and reduced cortical apoptosis in the basal cortex at 24 h after SAH. These findings indicated that TAT-mGluR1 acted against SAH-induced cell apoptosis through preventing mGluR1α truncation.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Receptores de Glutamato Metabotrópico/metabolismo , Hemorragia Subaracnoidea/metabolismo , Animales , Apoptosis/efectos de los fármacos , Calpaína/metabolismo , Masculino , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Hemorragia Subaracnoidea/inducido químicamente
19.
Brain Res Bull ; 137: 294-300, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29330035

RESUMEN

Emerging evidence indicates that loss of inhibitory tone in amygdala with its subsequent overactivation contributes to the development of multiple mental disorders such as anxiety disorders and post-traumatic stress disorder (PTSD). Harmine is a member of natural ß-carboline alkaloids which can readily cross the blood brain barrier and displays significant antidepressant and anxiolytic effects in rodents. However, the underlying neurobiological mechanisms are largely unknown. Here, by using whole-cell patch clamp recordings in in vitro amygdala slices, we examined the effect of harmine on glutamatergic and GABAergic transmission onto basal amygdala (BA) projection neurons (PNs). Our results showed that harmine affected neither the amplitude nor the frequency of spontaneous and miniature excitatory postsynaptic currents (sEPSCs/mEPSCs) of PNs. By contrast, it markedly increased both the amplitude and frequency of the spontaneous inhibitory postsynaptic currents (sIPSCs). For mIPSCs, only an increase of their frequency but not amplitude was observed following harmine perfusion, suggesting that harmine might act through presynaptic mechanism. In parallel, a reduction of paired-pulse ratio of evoked IPSCs emerged in the presence of harmine. Furthermore, the intrinsic excitability of PNs was dramatically decreased upon harmine treatment. Together, our study suggests that harmine selectively potentiates the inhibitory but not excitatory transmission onto BA PNs, which may contribute to its antidepressant and anxiolytic influence.


Asunto(s)
Ansiolíticos/farmacología , Complejo Nuclear Basolateral/efectos de los fármacos , Harmina/farmacología , Células Piramidales/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Animales , Complejo Nuclear Basolateral/metabolismo , Ácido Glutámico/metabolismo , Masculino , Ratones Endogámicos C57BL , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Técnicas de Placa-Clamp , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Células Piramidales/metabolismo , Transmisión Sináptica/fisiología , Técnicas de Cultivo de Tejidos
20.
Exp Neurol ; 301(Pt A): 13-25, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29258835

RESUMEN

Excessive glutamate in cerebrospinal fluid after subarachnoid hemorrhage (SAH) causes excitotoxic damage through calcium overloading and a subsequent apoptotic cascade. GluN1/GluN2B containing N-methyl-Daspartate (NMDA) receptor and metabotropic glutamate receptor 1 (mGluR1) can play a leading role in glutamate-mediated excitotoxicity. Here we report that Ifenprodil (100µM), a negative allosteric modulator (NAM) of GluN1/GluN2B NMDA receptors, and JNJ16259685 (10µM), a NAM of mGluR1, have an additive efficacy against glutamate (100µM)-induced Ca2+ release and cell apoptosis in primary cortical, hippocampal, and cerebellar granule neurons. Compared with intraperitoneal injection of Ifenprodil (10mg/kg) and JNJ16259685 (1mg/kg) separately, the combination therapy of Ifenprodil plus JNJ16259685 significantly improves the neurological deficit at 24h and 72h after experimental SAH. It reduces the number of TUNEL/DAPI-positive and activated caspase-3/NeuN-positive cells in cortical and hippocampal CA1 regions at 72h, decreases levels of glutamate in cerebrospinal fluid at 72h, and reduces the mitochondrial Ca2+ concentration. Meanwhile, the combination therapy attenuates apoptosis as shown by an increased Bcl-2 expression, decreased Bax expression and release of cytochrome c, and reduction of cleaved caspase-9 and caspase-3 at 24h after SAH. These findings indicate that targeting both the intracellular Ca2+ overloading and neuronal apoptosis using the Ifenprodil and JNJ16259685 is a promising new therapy for SAH.


Asunto(s)
Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Piperidinas/farmacología , Quinolinas/farmacología , Hemorragia Subaracnoidea , Antagonistas Adrenérgicos alfa/farmacología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Hemorragia Subaracnoidea/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...