Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Int J Mol Sci ; 24(17)2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37686174

ABSTRACT

Glioblastoma (GBM) is the most common primary brain malignancy in adults. Despite multimodal treatment that involves maximal safe resection, concurrent chemoradiotherapy, and tumour treatment for supratentorial lesions, the prognosis remains poor. The current median overall survival is only <2 years, and the 5-year survival is only 7.2%. Thioredoxin domain-containing protein 11 (TXNDC11), also known as EF-hand binding protein 1, was reported as an endoplasmic reticulum stress-induced protein. The present study aimed to elucidate the prognostic role of TXNDC11 in GBM. We evaluated the clinical parameters and TXNDC11 scores in gliomas from hospitals. Additionally, proliferation, invasion, migration assays, apoptosis, and temozolomide (TMZ)-sensitivity assays of GBM cells were conducted to evaluate the effects of short interfering RNA (siRNA) on these processes. In addition, these cells were subjected to Western blotting to detect the expression levels of N-cadherin, E-cadherin, and Cyclin D1. High levels of TXNDC11 protein expression were significantly associated with World Health Organization (WHO) high-grade tumour classification and poor prognosis. Multivariate analysis revealed that in addition to the WHO grade, TXNDC11 protein expression was also an independent prognostic factor of glioma. In addition, TXNDC11 silencing inhibited proliferation, migration, and invasion and led to apoptosis of GBM cells. However, over-expression of TXNDC11 enhanced proliferation, migration, and invasion. Further, TXNDC11 knockdown downregulated N-cadherin and cyclin D1 expression and upregulated E-cadherin expression in GBM cells. Knock-in TXNDC11 return these. Finally, in vivo, orthotopic xenotransplantation of TXNDC11-silenced GBM cells into nude rats promoted slower tumour growth and prolonged survival time. TXNDC11 is a potential oncogene in GBMs and may be an emerging therapeutic target.


Subject(s)
Glioblastoma , Glioma , Animals , Rats , Cadherins , Cyclin D1 , Glioma/genetics , Thioredoxins/genetics , Humans
2.
Environ Toxicol ; 38(9): 2143-2154, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37283489

ABSTRACT

Organophosphate pesticides (OPs), which are among the most widely used synthetic chemicals for the control of a wide variety of pests, are however associated with various adverse reactions in animals and humans. Chlorpyrifos, an OP, has been shown to cause various health complications due to ingestion, inhalation, or skin absorption. The mechanisms underlying the adverse effect of chlorpyrifos on neurotoxicity have not been elucidated. Therefore, we aimed to determine the mechanism of chlorpyrifos-induced cytotoxicity and to examine whether the antioxidant vitamin E (VE) ameliorated these cytotoxic effects using DBTRG-05MG, a human glioblastoma cell line. The DBTRG-05MG cells were treated with chlorpyrifos, VE, or chlorpyrifos plus VE and compared with the untreated control cells. Chlorpyrifos induced a significant decrease in cell viability and caused morphological changes in treated cultures. Furthermore, chlorpyrifos led to the increased production of reactive oxygen species (ROS) accompanied by a decrease in the level of reduced glutathione. Additionally, chlorpyrifos induced apoptosis by upregulating the protein levels of Bax and cleaved caspase-9/caspase-3 and by downregulating the protein levels of Bcl-2. Moreover, chlorpyrifos modulated the antioxidant response by increasing the protein levels of Nrf2, HO-1, and NQO1. However, VE reversed the cytotoxicity and oxidative stress induced by chlorpyrifos treatment in DBTRG-05MG cells. Overall, these findings suggest that chlorpyrifos causes cytotoxicity through oxidative stress, a process that may play an important role in the development of chlorpyrifos-associated glioblastoma.


Subject(s)
Antioxidants , Chlorpyrifos , Insecticides , Vitamin E , Animals , Humans , Antioxidants/pharmacology , Antioxidants/metabolism , Apoptosis , Chlorpyrifos/toxicity , Glioblastoma/drug therapy , Glioblastoma/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Vitamin E/pharmacology , Insecticides/toxicity , Cell Line, Tumor , Caspase 9/metabolism , Caspase 3/metabolism
3.
Toxicon ; 221: 106957, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36374720

ABSTRACT

Mycotoxins are secondary metabolites produced by various kinds of fungi that can induce disease in humans. The fungal species Penicillium expansum produces patulin (C7H6O4), a polyketide lactone mycotoxin found in fruits. Patulin is classified as noncarcinogen; however, recently, it has been associated with harmful effects on the central nervous system. Patulin's toxic action has been established in various brain models; however, its effect on human glioblastoma remains elusive. This study explores whether patulin induces cytotoxicity through oxidative stress in DBTRG-05MG human glioblastoma cells. This study also evaluates whether the antioxidant N-acetylcysteine (NAC) protects against patulin-induced cytotoxicity. In DBTRG-05MG cells, patulin concentration (10-60 µM) dependently induced cytotoxicity. Concerning oxidative stress, patulin (10 and 20 µM) increased the production of intracellular reactive oxygen species (ROS) but depleted reduced glutathione (GSH) contents and regulated the expressions of antioxidant-related proteins (Nrf2 and HO-1). Furthermore, patulin induced cytotoxicity via modulation of apoptosis-related protein expressions (Bax, cleaved caspase-9, and cleaved caspase-3). These cytotoxic responses were partially reversed via pretreatment with NAC (10 µM). In summary, these data help us understand the toxicology of patulin in human glioblastoma and evaluate whether NAC could clinically reduce patulin-affected brain damage.


Subject(s)
Glioblastoma , Patulin , Humans , Patulin/toxicity , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Antioxidants/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
4.
Quant Imaging Med Surg ; 12(1): 558-567, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34993101

ABSTRACT

BACKGROUND: Susceptibility-weighted imaging (SWI) is sensitive to the accumulation of paramagnetic substances, such as hemorrhage and increased venous vasculature, both being frequently found in high-grade tumors. The purpose of this retrospective study is to differentiate high-grade and low-grade astrocytoma by objectively measuring quantitative intra-tumoral susceptibility signals (qITSS) on SWI. METHODS: Precontrast SWI and 3D contrast-enhanced T1WI of 65 patients with astrocytoma were collected at 1.5 Tesla. All tumors were histologically confirmed and classified into two groups: high grade (WHO grade III and IV, n=50) and low grade (WHO grade II, n=15). After manual delineation of the tumor on T1WI, normalized contrast (NC) was calculated voxel by voxel within the tumor by using the concept of contrast to noise ratio. Thresholding on NC was applied to detect qITSS, and the volumetric percentage of qITSS can be obtained for each tumor. Two-sample t-test was applied to examine significant difference of qITSS percentage between high-grade and low-grade astrocytoma for different NC thresholds, ranging from 4 to 20. Receiver operating characteristic analysis was performed to evaluate the performance of differentiation. RESULTS: P value was less than 0.01 for a large range of NC thresholds [4-20], reflecting significant difference of qITSS percentage between high-grade and low-grade astrocytoma. The area under the receiver operating characteristic curve was larger than 0.9 at NC thresholds from 8 to 16 and peaks at 0.949 with a NC threshold of 14. It was shown that astrocytoma grading by qITSS percentage is successful for a wide range of NC threshold, demonstrating robustness on threshold selection. CONCLUSIONS: Without relying on the selection of slice position and at the same time providing objective identification of hypointense signal in SWI, the qITSS percentage can be used to distinguish high-grade and low-grade astrocytoma reliably.

5.
World Neurosurg ; 158: 205-209, 2022 02.
Article in English | MEDLINE | ID: mdl-34839047

ABSTRACT

OBJECTIVE: Ependymomas are rare central nervous system tumors. The current treatment strategy is gross total tumor removal. Whether adjuvant therapy will be beneficial is controversial. We retrospectively analyzed 3 cases of World Health Organization (WHO) grade III posterior fossa anaplastic ependymomas treated with different treatment modalities. We aimed to identify possible treatment options for infratentorial WHO grade III anaplastic ependymoma in adults. METHODS: We performed a retrospective analysis of 3 patients diagnosed with infratentorial anaplastic ependymomas in our institution from 2016 to 2020. The demographic data were documented. This case series of 3 patients does not meet the Department of Health and Human Services definition of research and does not need Institutional Review Board approval. All patients' informed consents have been obtained. RESULTS: One patient underwent subtotal tumor resection combined with adjuvant radiotherapy and Gamma Knife radiosurgery while the other 2 patients underwent gross total tumor removal combined with Gamma Knife radiosurgery or adjuvant radiotherapy. Tumors recurred in the first patient 20 months later, while the other 2 patents did not develop recurrence. The modified Rankin scale scores of these patients were 1, 0, and 0. All patients are followed up with regular magnetic resonance imaging at our facility. CONCLUSIONS: The strategy for treating WHO grade III anaplastic ependymomas is controversial, but gross total tumor resection remains the key element. Adjuvant stereotactic radiosurgery after tumor removal might be considered if radiotherapy is not an option. The role of chemotherapy is unclear, and the use of chemotherapy should be tailored to individual patients.


Subject(s)
Ependymoma , Infratentorial Neoplasms , Adult , Ependymoma/diagnostic imaging , Ependymoma/surgery , Humans , Infratentorial Neoplasms/diagnostic imaging , Infratentorial Neoplasms/surgery , Radiotherapy, Adjuvant , Retrospective Studies
6.
Pestic Biochem Physiol ; 179: 104979, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34802529

ABSTRACT

Rotenone, a plant-derived pesticide belonging to genera Derris and Lonchorcarpus, is an inhibitor of NADH dehydrogenase complex. Studies have shown that rotenone was applied as a neurotoxic agent in various neuronal models. Hydroxytyrosol [2-(3,4-dihydroxyphenyl)-ethanol] is a natural phenolic compound found in the olive (Olea europaea L.). Studies of hydroxytyrosol have dramatically increased because this compound may contribute to the prevention of neurodegenerative diseases. Although hydroxytyrosol has received increasing attention due to its multiple pharmacological activities, it is not explored whether hydroxytyrosol inhibited rotenone-induced cytotoxicity in the neuronal cell model. The aim of this study was to explore whether hydroxytyrosol prevented rotenone-induced Ca2+ signaling, cytotoxicity and oxidative stress in HCN-2 neuronal cell line. In HCN-2 cells, rotenone (5-30 µM) concentration-dependently induced cytosolic Ca2+ concentrations ([Ca2+]i) rises and cytotoxicity. Treatment with hydroxytyrosol (30 µM) reversed rotenone (20 µM)-induced cytotoxic responses. In Ca2+-containing medium, rotenone-induced Ca2+ entry was inhibited by 2-APB (a store-operated Ca2+ channel modulator) or hydroxytyrosol. In Ca2+-free medium, treatment with thapsigargin (an endoplasmic reticulum Ca2+ pump inhibitor) or hydroxytyrosol significantly inhibited rotenone-induced [Ca2+]i rises. Furthermore, treatment with hydroxytyrosol reversed ROS levels, cytotoxic responses, and antioxidant enzyme activities (SOD, GPX and CAT) in rotenone-treated cells. Together, in HCN-2 cells, rotenone induced Ca2+ influx via store-operated Ca2+ entry and Ca2+ release from the endoplasmic reticulum and caused oxidative stress. Moreover, hydroxytyrosol ameliorated Ca2+ or ROS-associated cytotoxicity. It suggests that hydroxytyrosol might have a protective effect on rotenone-induced neurotoxicity in human neuronal cells.


Subject(s)
Pesticides , Rotenone , Calcium/metabolism , Cell Survival , Oxidative Stress , Phenylethyl Alcohol/analogs & derivatives , Rotenone/toxicity
7.
J Biochem Mol Toxicol ; 35(10): e22862, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34309954

ABSTRACT

Cinobufagin, a bufadienolide of toad venom of Bufo bufo gargarizans, is used as a cardiotonic, central nervous system (CNS) respiratory agent, as well as an analgesic and anesthetic. However, several research showed that bufadienolide has a few side effects on the CNS, such as breathlessness or coma. Although cinobufagin was shown to display pharmacological effects in various models, the toxic effect of cinobufagin is elusive in brain cell models. The aim of this study was to explore whether cinobufagin affected viability, Ca2+ homeostasis, and reactive oxygen species (ROS) production in Gibco® Human Astrocyte (GHA) and HCN-2 neuronal cell line. In GHA cells but not in HCN-2 cells, cinobufagin (20-60 µM) induced [Ca2+ ]i rises. In terms of cell viability, chelation of cytosolic Ca2+ with 1,2-bis(2-aminophenoxy)ethane-N,N,N'N'-tetraacetic acid reduced cinobufagin-induced cytotoxicity in GHA cells. In GHA cells, cinobufagin-induced Ca2+ entry was inhibited by 2-aminoethoxydiphenyl borate or SKF96365. In a Ca2+ -free medium, treatment with thapsigargin or U73122 abolished cinobufagin-evoked [Ca2+ ]i rises. Furthermore, treatment with N-acetylcysteine reversed ROS production and cytotoxicity in cinobufagin-treated GHA cells. Together, in GHA cells but not in HCN-2 cells cinobufagin caused cytotoxicity that was linked to preceding [Ca2+ ]i rises by Ca2+ influx via store-operated Ca2+ entry and phospholipase C-dependent Ca2+ release from the endoplasmic reticulum. Moreover, cinobufagin induced ROS-associated cytotoxicity.


Subject(s)
Amphibian Venoms/chemistry , Astrocytes/metabolism , Brain/metabolism , Bufanolides/pharmacology , Calcium Signaling/drug effects , Homeostasis/drug effects , Neurons/metabolism , Reactive Oxygen Species/metabolism , Animals , Astrocytes/drug effects , Brain/pathology , Bufanolides/chemistry , Bufanolides/isolation & purification , Bufo bufo , Calcium/metabolism , Cell Line , Cell Survival/drug effects , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Neurons/drug effects , Thapsigargin/pharmacology , Type C Phospholipases/metabolism
8.
Environ Toxicol ; 36(7): 1375-1388, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33818898

ABSTRACT

Fusarium mycotoxins are one of the largest families of mycotoxins. Among these mycotoxins, deoxynivalenol is the most widespread pollutant of grains. However, the mechanism underlying the effect of deoxynivalenol on cytotoxicity in human brain endothelial cells was still unclear. This study examined whether deoxynivalenol induced oxidative stress-associated cytotoxicity in primary human brain endothelial cells (HBEC-5i), and explored whether Vitamin E (VE), a selective antioxidant, had protective effects on deoxynivalenol-treated cells. Deoxynivalenol (10-50 µM) concentration-dependently induced cytotoxicity in HBEC-5i cells. Deoxynivalenol (IC50 = 20 µM) activated mitochondrial apoptotic pathway by modulating antioxidant protein expressions (Nrf2, HO-1 and NQO1). More significantly, pre-treatment with VE (20 µM) attenuated the deoxynivalenol-induced cytotoxicity in this cell model. Together, VE significantly alleviated the apoptotic effects of deoxynivalenol in HBEC-5i cells suggesting that it protected the cells against deoxynivalenol-induced oxidative damage. Our findings provided new insight that VE had the potential to ameliorate neurotoxicity of deoxynivalenol.


Subject(s)
Mycotoxins , Vitamin E , Brain/metabolism , Endothelial Cells/metabolism , Humans , Mycotoxins/toxicity , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Reactive Oxygen Species , Trichothecenes , Vitamin E/pharmacology
9.
Clin Exp Pharmacol Physiol ; 48(5): 801-810, 2021 05.
Article in English | MEDLINE | ID: mdl-33609056

ABSTRACT

Hypaconitine, a neuromuscular blocker, is a diterpene alkaloid found in the root of Aconitum carmichaelii. Although hypaconitine was shown to affect various physiological responses in neurological models, the effect of hypaconitine on cell viability and the mechanism of its action of Ca2+ handling is elusive in cortical neurons. This study examined whether hypaconitine altered viability and Ca2+ signalling in HCN-2 neuronal cell lines. Cell viability was measured by the cell proliferation reagent (WST-1). Cytosolic Ca2+ concentrations [Ca2+ ]i was measured by the Ca2+ -sensitive fluorescent dye fura-2. In HCN-2 cells, hypaconitine (10-50 µmol/L) induced cytotoxicity and [Ca2+ ]i rises in a concentration-dependent manner. Removal of extracellular Ca2+ partially reduced the hypaconitine's effect on [Ca2+ ]i rises. Furthermore, chelation of cytosolic Ca2+ with BAPTA-AM reduced hypaconitine's cytotoxicity. In Ca2+ -containing medium, hypaconitine-induced Ca2+ entry was inhibited by modulators (2-APB and SKF96365) of store-operated Ca2+ channels and a protein kinase C (PKC) inhibitor (GF109203X). Hypaconitine induced Mn2+ influx indirectly suggesting that hypaconitine evoked Ca2+ entry. In Ca2+ -free medium, treatment with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin abolished hypaconitine-induced [Ca2+ ]i rises. Conversely, treatment with hypaconitine inhibited thapsigargin-induced [Ca2+ ]i rises. However, inhibition of phospholipase C (PLC) with U73122 did not inhibit hypaconitine-induced [Ca2+ ]i rises. Together, hypaconitine caused cytotoxicity that was linked to preceding [Ca2+ ]i rises by Ca2+ influx via store-operated Ca2+ entry involved PKC regulation and evoking PLC-independent Ca2+ release from the endoplasmic reticulum. Because BAPTA-AM loading only partially reversed hypaconitine-induced cell death, it suggests that hypaconitine induced a second Ca2+ -independent cytotoxicity in HCN-2 cells.


Subject(s)
Aconitine/analogs & derivatives , Egtazic Acid/analogs & derivatives , Calcium Signaling , Diterpene Alkaloids
10.
Neurotox Res ; 39(2): 256-265, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32588354

ABSTRACT

Mesaconitine, one of Aconitum carmichaelii Debx bioactive compounds, was shown to evoke Ca2+ homeostasis and its related physiological effects in endothelial cell types. However, the effect of mesaconitine on Ca2+ signaling and cell viability in human brain microvascular endothelial cells is unclear. This study focused on exploring whether mesaconitine changed cytosolic Ca2+ concentrations ([Ca2+]i), affected cell viability, and established the relationship between Ca2+ signaling and viability in HBEC-5i human brain microvascular endothelial cells. In HBEC-5i cells, cell viability was measured by the cell proliferation reagent (WST-1). [Ca2+]i was measured by the Ca2+-sensitive fluorescent dye fura-2. Mesaconitine (10-100 µM) concentration dependently induced [Ca2+]i rises. Ca2+ removal reduced the signal by approximately 25%. Mesaconitine (40-100 µM) caused cytotoxicity in HBEC-5i cells. This cytotoxic response was significantly reversed by chelation of cytosolic Ca2+ with BAPTA/AM. In Ca2+-containing medium, mesaconitine-induced Ca2+ entry was inhibited by 25% by modulators of store-operated Ca2+ channels and protein kinase C (PKC). Furthermore, mesaconitine also induced Mn2+ influx suggesting of Ca2+ entry. In Ca2+-free medium, treatment with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin abolished mesaconitine-evoked [Ca2+]i rises. Conversely, treatment with mesaconitine abolished thapsigargin-evoked [Ca2+]i rises. Inhibition of phospholipase C (PLC) with U73122 abolished mesaconitine-induced [Ca2+]i rises. In sum, mesaconitine caused cytotoxicity that was triggered by preceding [Ca2+]i rises. Furthermore, mesaconitine induced [Ca2+]i rises by evoking Ca2+ entry via PKC-sensitive store-operated Ca2+ channels and PLC-dependent Ca2+ release from the endoplasmic reticulum. It suggests that Ca2+ signaling have a potential cytotoxic effect on mesaconitine-treated human brain microvascular endothelial cells.


Subject(s)
Aconitine/analogs & derivatives , Calcium Signaling/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Aconitine/administration & dosage , Aconitum , Cell Line , Cell Survival/drug effects , Humans , Plant Extracts/toxicity
11.
Surg Neurol Int ; 11: 414, 2020.
Article in English | MEDLINE | ID: mdl-33365177

ABSTRACT

BACKGROUND: Intracranial solitary fibrous tumor/hemangiopericytoma (HPC) is a rare and aggressive tumor. We conducted this retrospective study to investigate the outcome of patients after treatment, the efficacy of postoperative adjuvant radiotherapy, and the factors not conducive to total resection. METHODS: We conducted a retrospective review of the medical records of patients harboring fresh intracranial solitary fibrous tumor/HPC treated from January 2009 to December 2019 in our hospital. We reviewed their clinical presentations, radiologic appearances, tumor size and location, extent of resection, estimate intraoperative blood loss, treatment modalities and results, and duration of follow-up. RESULTS: There were seven consecutive patients (three males and four females). The ages of the patients at the time of diagnosis ranged from 35 to 77 years (mean: 52.86 years). Five patients (71.43%) got tumor bigger than 5 cm in dimension and only 1 patient (14.29%) underwent gross total tumor resection in the first operation without complication. Five patients (71.43%) underwent postoperative adjuvant radiotherapy. Follow-up period ranged from 4.24 to 123.55 months and the median follow-up period was 91.36 months. Three patients had favorable outcome with Glasgow Outcome Scale (GOS) equal to 4; four patients had unfavorable outcome with GOS equal to 2 or 3. No mortality was happened. CONCLUSION: Gross total tumor resection in the initial surgery is very important to achieve a better outcome. Massive intraoperative bleeding and venous sinus or major vessels adjoining are factors not conducive to total resection. Radiotherapy can be administered as adjuvant therapy for cases showing an aggressive phenotype or not treated with gross total resection.

12.
Naunyn Schmiedebergs Arch Pharmacol ; 393(11): 2117-2127, 2020 11.
Article in English | MEDLINE | ID: mdl-32594194

ABSTRACT

Haloperidol, a typical antipsychotic medication, has been shown to possess various biological effects in different brain models. However, the impact of haloperidol on Ca2+ signaling in astrocytes is elusive. This study explored the effect of haloperidol on cytosolic free Ca2+ levels ([Ca2+]i) and viability, and established these two connections in Gibco® Human Astrocytes (GHAs) and DI TNC1 rat astrocytes. Haloperidol (5-20 µM) caused [Ca2+]i rises in a concentration-dependent manner in GHAs but not in DI TNC1 cells. Furthermore, removal of extracellular Ca2+ reduced haloperidol's effect by approximately 30% in GHAs. Haloperidol (20-40 µM) evoked concentration-dependent cytotoxicity in GHAs and DI TNC1 cells. However, chelating cytosolic Ca2+ with the Ca2+ chelator BAPTA/AM significantly reversed haloperidol's cytotoxicity only in GHAs. In GHAs, haloperidol-induced Ca2+ entry was inhibited by store-operated Ca2+ modulators (2-APB and SKF96365) and the protein kinase C (PKC) inhibitor GF109203X. This Ca2+ entry induced by haloperidol was confirmed by Mn2+ entry-induced quench of fura-2 fluorescence. In Ca2+-free medium, treatment with the endoplasmic reticulum Ca2+ pump inhibitor 2,5-di-tert-butylhydroquinone (BHQ) abolished haloperidol-induced [Ca2+]i rises. Conversely, treatment with haloperidol inhibited 45% of BHQ-evoked [Ca2+]i rises. Moreover, haloperidol-induced Ca2+ release from the endoplasmic reticulum was abolished by inhibition of phospholipase C (PLC) by U73122. Together, in GHAs but not in DI TNC1 cells, haloperidol caused Ca2+-associated cell death, induced Ca2+ entry via PKC-sensitive store-operated Ca2+ channels, and evoked PLC-dependent Ca2+ release from the endoplasmic reticulum. The protective effect of Ca2+ chelating on haloperidol-induced cytotoxicity in human astrocytes was also demonstrated.


Subject(s)
Antipsychotic Agents/toxicity , Astrocytes/drug effects , Calcium Chelating Agents/pharmacology , Calcium Signaling/drug effects , Egtazic Acid/analogs & derivatives , Haloperidol/toxicity , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Protein Kinase C/metabolism , Rats , Species Specificity , Type C Phospholipases/metabolism
13.
Eur J Radiol ; 117: 56-61, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31307653

ABSTRACT

PURPOSE: The purpose of this retrospective study was to investigate the differentiation of abscess and necrotic tumors, using susceptibility-weighted imaging (SWI) and apparent diffusion coefficients (ADC) either separated or combined. METHODS: Imaging was performed on 26 patients with pyogenic brain abscesses, 31 patients with rim-enhancing glioblastomas, and 21 patients with rim-enhancing metastases. The degree of intralesional susceptibility signal (ILSS) was independently assessed by three observers. Average ADC in the lesion core was calculated. After receiver operating characteristic (ROC) analysis, the area under the ROC curve was compared using three different analytical models (ILSS, ADC, and ILSS-ADC combined) to differentiate abscess from the two rim-enhancing necrotic tumors. RESULTS: The ILSS-ADC combined model had greater area under the ROC curves than ILSS or ADC used alone. In this study, the ILSS-ADC combined model showed 100% diagnostic accuracy differentiating abscesses from glioblastoma. The ADC model and the ILSS-ADC combined model performed equally well in distinguishing abscesses from metastases. CONCLUSION: It is concluded that SWI and ADC are complementary, and the combination of SWI and ADC may improve results compared with the use of only one model. Validation by an independent cohort is the next necessary step to broaden its applicability in routine clinical settings.


Subject(s)
Brain Abscess/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Glioblastoma/diagnostic imaging , Necrosis/diagnostic imaging , Adult , Aged , Brain Abscess/pathology , Brain Neoplasms/pathology , Diagnosis, Differential , Diffusion Magnetic Resonance Imaging/methods , Female , Glioblastoma/pathology , Humans , Male , Middle Aged , Necrosis/pathology , Retrospective Studies
14.
Pestic Biochem Physiol ; 157: 152-160, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31153463

ABSTRACT

Malathion, one of commonly used organophosphate insecticides, has a wide range of toxic actions in different models. However, the effect of this compound on Ca2+ homeostasis and its related cytotoxicity in glial cells is elusive. This study examined whether malathion evoked intracellular Ca2+ concentration ([Ca2+]i) rises and established the relationship between Ca2+ signaling and cytotoxicity in normal human astrocytes, rat astrocytes and human glioblastoma cells. The data show that malathion induced concentration-dependent [Ca2+]i rises in Gibco® Human Astrocytes (GHA cells), but not in DI TNC1 normal rat astrocytes and DBTRG-05MG human glioblastoma cells. In GHA cells, this Ca2+ signal response was reduced by removing extracellular Ca2+. In Ca2+-free medium, pretreatment with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin abolished malathion-induced [Ca2+]i rises. Conversely, incubation with malathion abolished thapsigargin-induced [Ca2+]i rises. Inhibition of phospholipase C (PLC) with U73122 also blocked malathion-induced [Ca2+]i rises. In Ca2+-containing medium, malathion-induced [Ca2+]i rises was inhibited by store-operated Ca2+ channel blockers (2-APB, econazole or SKF96365) and the protein kinase C (PKC) inhibitor GF109203X. Malathion (5-25 µM) concentration-dependently caused cytotoxicity in GHA, DI TNC1 and DBTRG-05MG cells. This cytotoxic effect was partially prevented by prechelating cytosolic Ca2+ with BAPTA-AM (a selective Ca2+ chelator) only in GHA cells. Together, in GHA but not in DI TNC1 and DBTRG-05MG cells, malathion induced [Ca2+]i rises by inducing PLC-dependent Ca2+ release from the endoplasmic reticulum and Ca2+ entry via PKC-sensitive store-operated Ca2+ channels. Furthermore, malathion induced Ca2+-associated cytotoxicity, suggesting that Ca2+ chelating may have a protective effect on malathion-induced cytotoxicity in normal human astrocytes.


Subject(s)
Calcium/metabolism , Malathion/pharmacology , Animals , Calcium Signaling/drug effects , Cell Line , Cell Survival/drug effects , Chelating Agents , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Neuroglia/drug effects , Neuroglia/metabolism , Rats
15.
Neurotoxicology ; 69: 97-107, 2018 12.
Article in English | MEDLINE | ID: mdl-30292652

ABSTRACT

Exposure to insecticides has been found to have deleterious effects on human health. Lambda-cyhalothrin (LCT), a mixture of isomers of cyhalothrin, is a pyrethroid insecticide routinely used in pest control programs. LCT was reported to cause neurotoxic effects in various models. However, the mechanism of underlying effect of LCT on cytotoxicity in normal human brain cells is still elusive. This study examined whether LCT affected Ca2+ homeostasis and Ca2+-related physiology in Gibco® Human Astrocytes (GHA cells), and explored whether BAPTA-AM (1,2-bis(2-aminophenoxy)ethane-N,N,N'N'-tetraacetic acid), a selective Ca2+ chelator, has protective effects on LCT-treated GHA cells. The data show that LCT (10-15 µM) concentration-dependently induced cytotoxicity in both GHA cells and DI TNC1 normal rat astrocytes but only induced intracellular Ca2+ concentration ([Ca2+]i) rises in GHA cells. In terms of Ca2+ signaling in GHA cells, LCT-induced [Ca2+]i rises were reduced by removing extracellular Ca2+ and were inhibited by store-operated Ca2+ channel modulators (2-APB, econazole or SKF96365). In Ca2+-free medium, pretreatment with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin abolished LCT-induced [Ca2+]i rises. Conversely, incubation with LCT abolished thapsigargin-induced [Ca2+]i rises. Regarding cytotoxicity, LCT evoked apoptosis by regulating apoptotic protein expressions (Bax, BCl-2, cleaved caspase-9/-3). This apoptotic response was significantly inhibited by prechelating cytosolic Ca2+ with BAPTA-AM. Together, in GHA cells, LCT induced [Ca2+]i rises by inducing Ca2+ entry via store-operated Ca2+ channels and Ca2+ release from the endoplasmic reticulum. Moreover, BAPTA-AM has a protective effect on inhibiting LCT-activated mitochondrial apoptotic pathway. This study provided new insights into the molecular protective mechanism of LCT-induced cytotoxicity in normal human astrocytes.


Subject(s)
Astrocytes/drug effects , Calcium Signaling/drug effects , Egtazic Acid/analogs & derivatives , Insecticides/toxicity , Mitochondria/drug effects , Nitriles/toxicity , Pyrethrins/toxicity , Animals , Apoptosis/drug effects , Apoptosis/physiology , Astrocytes/metabolism , Calcium/metabolism , Calcium Chelating Agents/pharmacology , Calcium Signaling/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Humans , Mitochondria/metabolism , Rats
16.
Case Rep Surg ; 2018: 1382680, 2018.
Article in English | MEDLINE | ID: mdl-29992076

ABSTRACT

INTRODUCTION: Glioblastoma multiforme (GBM), the most common primary malignant brain tumor in adults, is characterized by extensive heterogeneity in its clinicopathological presentation. A primary brain tumor with both astrocytic differentiation and neuronal immunophenotype features is rare. Here, we report a long-term survival patient who presented this rare form of GBM in the disease course. PRESENTATION OF CASE: A 23-year-old woman, presenting with rapidly progressive headache and right-side weakness, was diagnosed with brain tumor over the left basal ganglion. She underwent the first craniectomy for tumor removal, and histopathology revealed classic GBM. Tumor recurrence occurred 8 years later. Another gross total resection was performed and pathology revealed GBM with the oligodendroglioma component (GBM-O). Due to disease progression, she received debulking surgery the following year. The third pathology revealed glioblastoma with primitive neuroectodermal tumor-like component (GBM-PNET). DISCUSSION: GBM-PNETs are collision tumors with both neuronal and glial components. They are rare, and a few case reports have suggested that these tumors are associated with favorable outcomes but a higher risk of cerebrospinal fluid dissemination. CONCLUSION: We report a patient who developed the distinct pathologic variants of classic GBM, GBM-O, and GBM-PNET, throughout the disease course. Young age, aggressive surgical resection, and pathologic and genetic features may have contributed to the long-term survival of the patient.

17.
Surg Neurol Int ; 9: 127, 2018.
Article in English | MEDLINE | ID: mdl-30034918

ABSTRACT

BACKGROUND: Giant cell tumor of bone originating from the connective tissue within the bone marrow is benign but locally aggressive lesion. In all, 90% of the cases involve the epiphysis of long bones and less than 2% involve the skull. Giant cell tumors of the skull occur most frequently in the sphenoid and temporal bones, and very rarely in the ethmoid, frontal, parietal, and occipital bones. We would like to share a case of giant cell tumor of bone arising from the left orbital roof with involving ethmoid sinus, which was diagnosed to be a meningioma before surgery. CASE DESCRIPTION: A 32-year-old lady presented to us with the chief complain of left proptosis, diplopia, and left eye soreness without decline of visual acuity for about 2 months. Her orbital magnetic resonance imaging (MRI) disclosed a mass lesion located in the left frontal base, orbital roof, and upper medial orbital region with adjacent dural-tail sign favoring meningioma. She underwent a left supraorbital pterional craniotomy with the gross total removal of tumor and dura reconstruction. Histology examination of the tumor showed a picture of giant cell tumor of bone. Considering giant cell tumor of bone is locally aggressive, postoperative adjuvant therapy with Denosumab was introduced after full explanation. CONCLUSION: Standard treatments of skull-base giant cell tumors have yet to be established due to small number of cases reported in the literature. The standard treatment of giant cell tumor of bone is complete resection of the tumor.

18.
Environ Toxicol ; 32(12): 2440-2454, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28804952

ABSTRACT

Propofol (2,6-diisopropylphenol), one of the extensively and commonly used anesthetic agents, has been shown to affect the biological behavior of various models. Previous researches have shown that propofol-induced cytotoxicity might cause anticancer effect in different cells. However, the mechanisms underlying the effect of propofol on cytotoxicity is still elusive in human glioblastoma cells. The aims of this study were to evaluate effects of propofol on cytotoxicity, cell cycle distribution and ROS production, and establish the relationship between oxidative stress and cytotoxicity in GBM 8401 human glioblastoma cells and DI TNC1 rat astrocytes. Propofol (20-30 µM) concentration-dependently induced cytotoxicity, cell cycle arrest, and increased ROS production in GBM 8401 cells but not in DI TNC1 cells. In GBM 8401 cells, propofol induced G2/M phase cell arrest, which affected the CDK1, cyclin B1, p53, and p21 protein expression levels. Furthermore, propofol induced oxygen stresses by increasing O2- and H2 O2 levels but treatment with the antioxidant N-acetylcysteine (NAC) partially reversed propofol-regulated antioxidative enzyme levels (superoxide dismutase, catalase, and glutathione peroxidase). Most significantly, propofol induced apoptotic effects by decreasing Bcl-2 but increasing Bax, cleaved caspase-9/caspase-3 levels, which were partially reversed by NAC. Moreover, the pancaspase inhibitor Z-VAD-FMK also partially prevented propofol-induced apoptosis. Together, in GBM 8401 cells but not in DI TNC1 cells, propofol activated ROS-associated apoptosis that involved cell cycle arrest and caspase activation. These findings indicate that propofol not only can be an anesthetic agent which reduces pain but also has the potential to be used for the treatment of human glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Astrocytes/drug effects , Propofol/pharmacology , Acetylcysteine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Astrocytes/cytology , Caspase 3/metabolism , Caspase 9/metabolism , Caspase Inhibitors/pharmacology , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Glioblastoma , Humans , Rats , Reactive Oxygen Species/metabolism
19.
Food Chem Toxicol ; 107(Pt A): 280-292, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28689919

ABSTRACT

Researches have been conducted to explore the biological effect of gastrodin, a natural compound extracted from the rhizome of Gastrodia elata Blume, in different models. However, the effects of gastrodin on cytotoxicity, cell cycle distribution and oxidative stress in glia cells have not been explored. The aim of this study was to investigate the cytotoxic effect of gastrodin and its mechanisms in DBTRG-05MG human glioblastoma cells and CTX TNA2 rat astrocytes. In DBTRG-05MG cells but not in CTX TNA2 cells, gastrodin (20-30 µM) induced cytotoxicity, G2/M phase cell cycle arrest and apoptosis. Regarding oxidative stress, gastrodin (20-30 µM) elevated intracellular ROS levels but reduced GSH levels. Treatment with the antioxidant NAC (10 µM) partially reversed gastrodin-altered antioxidant enzymes levels. Furthermore, gastrodin induced mitochondria-associated apoptosis. The apoptotic effects evoked by gastrodin were partially inhibited by the antioxidant NAC and the pancaspase inhibitor Z-VAD-FMK. Together, in DBTRG-05MG cells, but not in CTX TNA2 cells, gastrodin activated ROS-associated mitochondrial apoptotic pathways that involved cell cycle arrest. These data provide insight into the molecular mechanisms governing the ability of gastrodin to induce cytotoxicity in human glioblastoma cells and further suggest that gastrodin is a new potential agent for the treatment of human gliblasoma.


Subject(s)
Apoptosis/drug effects , Benzyl Alcohols/pharmacology , Cell Cycle Checkpoints/drug effects , Gastrodia/chemistry , Glioblastoma/drug therapy , Glucosides/pharmacology , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/physiopathology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Rats , Reactive Oxygen Species/metabolism , Rhizome/chemistry , Tumor Suppressor Protein p53/genetics
20.
J Biochem Mol Toxicol ; 31(10)2017 Oct.
Article in English | MEDLINE | ID: mdl-28658523

ABSTRACT

Tetramethylpyrazine (TMP) is a compound purified from herb. Its effect on Ca2+ concentrations ([Ca2+ ]i ) in renal cells is unclear. This study examined whether TMP altered Ca2+ signaling in Madin-Darby canine kidney (MDCK) cells. TMP at 100-800 µM induced [Ca2+ ]i rises, which were reduced by Ca2+ removal. TMP induced Mn2+ influx implicating Ca2+ entry. TMP-induced Ca2+ entry was inhibited by 30% by modulators of protein kinase C (PKC) and store-operated Ca2+ channels. Treatment with the endoplasmic reticulum Ca2+ pump inhibitor 2,5-di-tert-butylhydroquinone (BHQ) inhibited 93% of TMP-evoked [Ca2+ ]i rises. Treatment with TMP abolished BHQ-evoked [Ca2+ ]i rises. Inhibition of phospholipase C (PLC) abolished TMP-induced responses. TMP at 200-1000 µM decreased viability, which was not reversed by pretreatment with the Ca2+  chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester. Together, in MDCK cells, TMP induced [Ca2+ ]i rises by evoking PLC-dependent Ca2+ release from endoplasmic reticulum and Ca2+ entry via PKC-sensitive store-operated Ca2+ entry. TMP also caused Ca2+ -independent cell death.


Subject(s)
Calcium Signaling/drug effects , Kidney Tubules/metabolism , Pyrazines/pharmacology , Animals , Cell Survival/drug effects , Dogs , Kidney Tubules/cytology , Madin Darby Canine Kidney Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...