Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Life (Basel) ; 14(1)2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38255754

ABSTRACT

A 50-year-old male presented to the emergency room after experiencing sudden right upper limb facial numbness and dysphasia, followed by full recovery. A brain CT scan showed hyperdense lesions within the left hemispheric sulcus, which raised suspicion of spontaneous subarachnoid hemorrhage. A T1-weighted MRI showed multiple tiny leptomeningeal enhancements in the same area, and a digital subtraction angiography showed no signs of vascular abnormality. Cerebrospinal fluid cytology revealed atypical melanin-containing cells with minimal pleomorphism. One month later, the patient developed sixth nerve palsy, which was determined to be due to intracranial hypertension. Multiple giant nevi on the legs, trunk, and scalp were also observed. A skin biopsy showed well-defined and symmetrical proliferation of melanocytic nevus cell nests in the dermis. An open biopsy was performed due to the suspicious leptomeningeal lesions, which surprisingly revealed diffuse and thick black-colored tissue infiltration of the leptomeninges. Pathology confirmed the diagnosis of meningeal melanocytosis. A ventriculoperitoneal shunt was then placed, and the patient's neurological symptoms gradually improved. Based on the presence of multiple giant nevi on the patient's skin and the finding of diffuse meningeal melanocytosis during the open biopsy, the patient was diagnosed with neurocutaneous melanosis. The patient received 6 cycles triweekly of Ipilimumab and Nivolumab 8 months after initial diagnosis. Unfortunately, the disease progressed and the patient passed away 14 months after initial diagnosis.

2.
Chem Biol Interact ; 382: 110645, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37482209

ABSTRACT

Neuroblastoma, the most common childhood tumor, are highly malignant and fatal because neuroblastoma cells extremely defend against apoptotic targeting. Traditional treatments for neuroblastomas are usually ineffective and lead to serious side effects and poor prognoses. In this study, we investigated the molecular mechanisms of resveratrol-induced insults to neuroblastoma cells and survival extension of nude mice with neuroblastomas, especially in the endoplasmic reticular (ER) stress-intracellular reactive oxygen species (iROS) axis-mediated signals. Resveratrol specifically killed neuroblastoma cells mainly via apoptosis and autophagy rather than necrosis. As to the mechanisms, resveratrol time-dependently triggered productions of Grp78 protein and iROS in neuroblastoma cells. Attenuating the ER stress-iROS signaling axis significantly suppressed resveratrol-induced autophagy, DNA damage, and cell apoptosis. Successively, resveratrol decreased phosphorylation of retinoblastoma protein and induced cell cycle arrest at the S phase, translocation of Bak protein to mitochondria, a reduction in the mitochondrial membrane potential, cascade activation of caspases-9, -3, and -6, and DNA fragmentation. Moreover, weakening the ER stress-iROS axis concomitantly overcome resveratrol-induced decreases in translocation of Rho protein to membranes and succeeding cell migration. Interestingly, administration of resveratrol did not cause significant side effects but could protect the neuroblastoma-bearing nude mice from body weight loss and consequently extended the animal survival. In parallel, resveratrol elevated levels of Grp78 and then induced cell apoptosis in neuroblastoma tissues. This study has shown that resveratrol could kill neuroblastoma cells and extend survival of animals with neuroblastomas by triggering the ER stress-iROS-involved intrinsic apoptosis and suppression of Rho-dependent cell migration. Our results imply the potential of resveratrol as a drug candidate for chemotherapy of neuroblastoma patients.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Neuroblastoma , Animals , Mice , Resveratrol/pharmacology , Resveratrol/therapeutic use , Mice, Nude , Apoptosis , Neuroblastoma/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress
3.
Front Neurol ; 14: 1149236, 2023.
Article in English | MEDLINE | ID: mdl-37213896

ABSTRACT

Managing acute innominate artery (IA) dissection associated with severe stenosis is challenging due to its rarity, possible complex dissection patterns, and compromised blood flow to the brain and upper extremities. This report describes our treatment strategy for this challenging disease using the kissing stent technique. A 61-year-old man had worsening of an acute IA dissection secondary to an extension of a treated aortic dissection. Four possible treatment strategies for kissing stent placement were proposed based on different approaches (open surgical or endovascular) and accesses (trans-femoral, trans-brachial, or trans-carotid access). We chose to place two stents simultaneously via a percutaneous retrograde endovascular approach through the right brachial artery and a combined open surgical distal clamping of the common carotid artery with a retrograde endovascular approach through the carotid artery. This hybrid approach strategy highlights the three key points for maintaining safety and efficacy: (1) good guiding catheter support is obtainable through retrograde, rather than antegrade, access to the lesion, (2) concomitant cerebral and upper extremity reperfusion is guaranteed by placing kissing stents into the IA, and (3) peri-procedural cerebral emboli are prevented by surgical exposure of the common carotid artery with distal clamping.

4.
J Exp Clin Cancer Res ; 41(1): 220, 2022 Jul 13.
Article in English | MEDLINE | ID: mdl-35831908

ABSTRACT

BACKGROUND: The mechanism by which glioblastoma evades temozolomide (TMZ)-induced cytotoxicity is largely unknown. We hypothesized that mitochondria plays a role in this process. METHODS: RNA transcriptomes were obtained from tumor samples and online databases. Expression of different proteins was manipulated using RNA interference or gene amplification. Autophagic activity and mitochondrial metabolism was assessed in vitro using the respective cellular and molecular assays. In vivo analysis were also carried out in this study. RESULTS: High SH3GLB1 gene expression was found to be associated with higher disease grading and worse survival profiles. Single-cell transcriptome analysis of clinical samples suggested that SH3GLB1 and the altered gene levels of oxidative phosphorylation (OXPHOS) were related to subsets expressing a tumor-initiating cell signature. The SH3GLB1 protein was regulated by promoter binding with Sp1, a factor associated with TMZ resistance. Downregulation of SH3GLB1 resulted in retention of TMZ susceptibility, upregulated p62, and reduced LC3B-II. Autophagy inhibition by SH3GLB1 deficiency and chloroquine resulted in attenuated OXPHOS expression. Inhibition of SH3GLB1 in resistant cells resulted in alleviation of TMZ-enhanced mitochondrial metabolic function, such as mitochondrial membrane potential, mitochondrial respiration, and ATP production. SH3GLB1 modulation could determine tumor susceptibility to TMZ. Finally, in animal models, resistant tumor cells with SH3GLB1 knockdown became resensitized to the anti-tumor effect of TMZ, including the suppression of TMZ-induced autophagy and OXPHOS. CONCLUSIONS: SH3GLB1 promotes TMZ resistance via autophagy to alter mitochondrial function. Characterizing SH3GLB1 in glioblastoma may help develop new therapeutic strategies against this disease in the future.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Autophagy , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Mitochondria , Temozolomide/pharmacology , Temozolomide/therapeutic use
5.
J Clin Med ; 10(24)2021 Dec 13.
Article in English | MEDLINE | ID: mdl-34945132

ABSTRACT

Compared to nonaggressive cranial dural arteriovenous fistulae (cDAVF), aggressive cDAVF carries leptomeningeal venous drainage (LVD) and has approximately 15% annual risk of hemorrhagic and non-hemorrhagic aggressive neurological presentations. In terms of aggressive clinical presentations, the previous classification does not adequately differentiate the higher risk group from the lower risk group. Herein, we retrospectively collected a series of patients with aggressive cDAVF and explored the risk factors for differentiating the higher-risk group from the lower-risk group with aggressive clinical presentations. We retrospectively collected patients with aggressive cDAVF from March 2011 to March 2019. The risk of aggressive clinical presentation was recorded. Risk factors were included in the analysis for aggressive clinical presentations. From March 2011 to March 2019, 37 patients had aggressive cDAVF. Among them, 24 presented with aggressive clinical presentation (20, hemorrhagic presentation; four, non-hemorrhagic presentation). In patients presenting with hemorrhage, four patients experienced early rebleeding after diagnosis. In the univariate analysis, risk location, directness of LVD, exclusiveness of LVD, and venous strain were significantly different in patients with aggressive clinical presentation. In the multivariate analysis, exclusiveness of LVD and venous strain were observed, with a significant difference between patients with aggressive clinical presentation and those with benign clinical presentation. Among patients with angiographically aggressive cDAVFs, approximately 65% presented with aggressive clinical presentations in our series. Among all potential risk factors, patients with exclusiveness of LVD and venous strain have even higher risk and should be treated aggressively and urgently.

6.
J Surg Case Rep ; 2021(8): rjab334, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34405041

ABSTRACT

Vertebro-venous fistula (VVF) refers to an abnormal arteriovenous shunt connecting the extracranial vertebral artery and the paraspinal venous structures. Coil embolization is the mainstay treatment of choice for VVF, and accurate definition of the endovascular target is mandatory. Traditionally, catheter-based angiograms are used for treatment planning, but those images lack bony information to delineate the precise relationship of the drainage veins to the spinal structure. Herein, we presented two VVF cases and demonstrated how we used intra-arterial cone-beam computed tomography angiography (IA-CBCTA) to determine the safe embolization zone for dense coil packing. We propose that IA-CBCTA is a useful adjunct in the endovascular planning of VVF by offering an image consisting of bony and vascular information.

7.
Oxid Med Cell Longev ; 2021: 5558618, 2021.
Article in English | MEDLINE | ID: mdl-34136065

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive brain tumor. Drug resistance mainly drives GBM patients to poor prognoses because drug-resistant glioblastoma cells highly defend against apoptotic insults. This study was designed to evaluate the effects of cobalt chloride (CoCl2) on hypoxic stress, autophagy, and resulting apoptosis of human and mouse drug-resistant glioblastoma cells. Treatment of drug-resistant glioblastoma cells with CoCl2 increased levels of hypoxia-inducible factor- (HIF-) 1α and triggered hypoxic stress. In parallel, the CoCl2-induced hypoxia decreased mitochondrial ATP synthesis, cell proliferation, and survival in chemoresistant glioblastoma cells. Interestingly, CoCl2 elevated the ratio of light chain (LC)3-II over LC3-I in TMZ-resistant glioblastoma cells and subsequently induced cell autophagy. Analyses by loss- and gain-of-function strategies further confirmed the effects of the CoCl2-induced hypoxia on autophagy of drug-resistant glioblastoma cells. Furthermore, knocking down HIF-1α concurrently lessened CoCl2-induced cell autophagy. As to the mechanisms, the CoCl2-induced hypoxia decreased levels of phosphoinositide 3-kinase (PI3K) and successive phosphorylations of AKT and mammalian target of rapamycin (mTOR) in TMZ-resistant glioblastoma cells. Interestingly, long-term exposure of human chemoresistant glioblastoma cells to CoCl2 sequentially triggered activation of caspases-3 and -6, DNA fragmentation, and cell apoptosis. However, pretreatment with 3-methyladenine, an inhibitor of autophagy, significantly attenuated the CoCl2-induced autophagy and subsequent apoptotic insults. Taken together, this study showed that long-term treatment with CoCl2 can induce hypoxia and subsequent autophagic apoptosis of drug-resistant glioblastoma cells via targeting the PI3K-AKT-mTOR pathway. Thus, combined with traditional prescriptions, CoCl2-induced autophagic apoptosis can be clinically applied as a de novo strategy for therapy of drug-resistant GBM patients.


Subject(s)
Brain Neoplasms/complications , Cell Hypoxia/genetics , Cobalt/adverse effects , Glioblastoma/complications , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Apoptosis , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Glioblastoma/pathology , Humans , Mice , Signal Transduction
8.
Life Sci ; 258: 118195, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32781073

ABSTRACT

AIMS: The estrogen-ERα axis participates in osteoblast maturation. This study was designed to further evaluated the roles of the estrogen-ERα axis in bone healing and the possible mechanisms. MAIN METHODS: Female ICR mice were created a metaphyseal bone defect in the left femurs and administered with methylpiperidinopyrazole (MPP), an inhibitor of ERα. Bone healing was evaluated using micro-computed tomography. Colocalization of ERα with alkaline phosphatase (ALP) and ERα translocation to mitochondria were determined. Levels of ERα, ERß, PECAM-1, VEGF, and ß-actin were immunodetected. Expression of chromosomal Runx2, ALP, and osteocalcin mRNAs and mitochondrial cytochrome c oxidase (COX) I and COXII mRNAs were quantified. Angiogenesis was measured with immunohistochemistry. KEY FINDINGS: Following surgery, the bone mass was time-dependently augmented in the bone-defect area. Simultaneously, levels of ERα were specifically upregulated and positively correlated with bone healing. Administration of MPP to mice consistently decreased levels of ERα and bone healing. As to the mechanisms, osteogenesis was enhanced in bone healing, but MPP attenuated osteoblast maturation. In parallel, expressions of osteogenesis-related ALP, Runx2, and osteocalcin mRNAs were induced in the injured zone. Treatment with MPP led to significant inhibition of the alp, runx2, and osteocalcin gene expressions. Remarkably, administration of MPP lessened translocation of ERα to mitochondria and expressions of mitochondrial energy production-related coxI and coxII genes. Furthermore, exposure to MPP decreased levels of PECAM-1 and VEGF in the bone-defect area. SIGNIFICANCE: The present study showed the contributions of the estrogen-ERα axis to bone healing through stimulation of energy production, osteoblast maturation, and angiogenesis.


Subject(s)
Bone Regeneration , Cell Differentiation , Energy Metabolism , Estrogen Receptor alpha/metabolism , Neovascularization, Physiologic , Osteoblasts/cytology , Signal Transduction , Alkaline Phosphatase/metabolism , Animals , Body Weight/drug effects , Bone Regeneration/drug effects , Bony Callus/drug effects , Bony Callus/pathology , Cell Differentiation/drug effects , Chromosomes, Mammalian/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Energy Metabolism/drug effects , Female , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Neovascularization, Physiologic/drug effects , Organ Size/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteogenesis/drug effects , Protein Transport/drug effects , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Up-Regulation/drug effects , Wound Healing/drug effects
9.
Molecules ; 25(12)2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32580515

ABSTRACT

An estrogen deficiency is the main cause of osteoporosis in postmenopausal women. In bone remodeling, estrogen receptors (ERs) can mediate estrogen-transducing signals. Methylpiperidinopyrazole (MPP) is a highly specific antagonist of ER-alpha (ERα). This study was designed to evaluate the effects of MPP on estrogen-induced energy production, subsequent osteoblast maturation, and the possible mechanisms. Exposure of primary osteoblasts isolated from neonatal rat calvarias to MPP did not affect cell morphology or survival. Estradiol can induce translocation of ERα into mitochondria from the cytoplasm. Interestingly, pretreatment of rat calvarial osteoblasts with MPP lowered estrogen-induced ERα translocation. Sequentially, estrogen-triggered expressions of mitochondrial energy production-linked cytochrome c oxidase (COX) I and COX II messenger (m)RNAs were inhibited following pretreatment with MPP. Consequently, MPP caused decreases in estrogen-triggered augmentation of the activities of mitochondrial respiratory complex enzymes and levels of cellular adenosine phosphate (ATP). During progression of osteoblast maturation, estrogen induced bone morphogenetic protein (BMP)-6 and type I collagen mRNA expressions, but MPP treatment inhibited such induction. Consequently, estrogen-induced osteoblast activation and mineralization were attenuated after exposure to MPP. Taken together, MPP suppressed estrogen-induced osteoblast maturation through decreasing chromosomal osteogenesis-related BMP-6 and type I collagen mRNA expressions and mitochondrial ATP synthesis due to inhibiting energy production-linked COX I and II mRNA expressions. MPP can appropriately be applied to evaluate estrogen-involved bioenergetics and osteoblast maturation.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogens/genetics , Osteoporosis/drug therapy , Pyrazoles/pharmacology , Animals , Bone Morphogenetic Protein 6/genetics , Cell Differentiation/drug effects , Electron Transport Complex IV/genetics , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Estrogens/metabolism , Female , Gene Expression Regulation/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Osteoblasts/drug effects , Osteocalcin/genetics , Osteogenesis/drug effects , Osteoporosis/metabolism , Osteoporosis/pathology , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Rats , Signal Transduction/drug effects
10.
BMC Neurol ; 20(1): 178, 2020 May 11.
Article in English | MEDLINE | ID: mdl-32393192

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is one of the most aggressive malignant brain tumors. Intracranial GBM metastases to the spine are rarely detected clinically. Secondary gliosarcomas after treatment of primary GBM are rarely described. CASE PRESENTATION: Herein, we report the case of a 53-year-old woman who presented to our emergency room with progressive headache and weakness on the left side. Plain computed tomography and contrast magnetic resonance imaging of the brain revealed an approximately 6.8 cm × 4.5 cm right temporoparietooccipital intraaxial cystic tumor with surrounding diffuse perifocal edema that caused midline shift toward the left. Emergency craniotomy was performed to remove the tumor, and pathological examination revealed GBM. The patient received proton beam therapy, Gliadel implantation, and oral temozolomide chemotherapy as well as targeted therapy with bevacizumab. Approximately 15 months after diagnosis, she underwent surgical resection of the right temporal recurrent tumor and was newly diagnosed as having a metastatic spinal tumor. Pathologically, the right temporal and metastatic spinal tumors were gliosarcoma and GBM, respectively. CONCLUSIONS: Concurrent spinal metastasis and gliosarcomatous transformation, which are two types of GBM complications, are rare. To our knowledge, this is the first report of a case of recurrent GBM with gliosarcoma after proton bean therapy.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Glioblastoma/secondary , Gliosarcoma/pathology , Neoplasm Recurrence, Local/pathology , Spinal Neoplasms/secondary , Antineoplastic Agents, Alkylating/therapeutic use , Brain/diagnostic imaging , Brain/surgery , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Craniotomy , Fatal Outcome , Female , Glioblastoma/diagnostic imaging , Glioblastoma/therapy , Humans , Magnetic Resonance Imaging , Middle Aged , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/therapy , Proton Therapy , Spinal Neoplasms/diagnostic imaging , Spinal Neoplasms/surgery , Temozolomide/therapeutic use , Tomography, X-Ray Computed
11.
Pharmaceutics ; 12(5)2020 May 24.
Article in English | MEDLINE | ID: mdl-32456305

ABSTRACT

Malignant glioma (MG) is extremely aggressive and highly resistant to chemotherapeutic agents. Using electrospraying, the potent chemotherapeutic agent 7-ethyl-10-hydroxycamptothecia (SN-38) was embedded into 50:50 biodegradable poly[(d,l)-lactide-co-glycolide] (PLGA) microparticles (SMPs). The SMPs were stereotactically injected into the brain parenchyma of healthy rats and intratumorally injected into F98 glioma-bearing rats for estimating the pharmacodynamics and therapeutic efficacy. SN-38 was rapidly released after injection and its local (brain tissue) concentration remained much higher than that in the blood for more than 8 weeks. Glioma-bearing rats were divided into three groups-group A (n = 13; stereotactically injected pure PLGA microparticles), group B (n = 12; stereotactically injected Gliadel wafer and oral temozolomide), and group C (n = 13; stereotactic and intratumoral introduction of SMPs). The SMPs exhibited significant therapeutic efficacy, with prolonged survival, retarded tumor growth, and attenuated malignancy. The experimental results demonstrated that SMPs provide an effective and potential strategy for the treatment of MG.

12.
Ther Adv Med Oncol ; 11: 1758835919875555, 2019.
Article in English | MEDLINE | ID: mdl-31632467

ABSTRACT

BACKGROUND: Malignant gliomas (MGs) are highly chemotherapy-resistant. Temozolomide (TMZ) and carmustine (BiCNU) are alkylating agents clinically used for treating MGs. However, their effectiveness is restrained by overexpression of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) in tumors. O6-benzylguanine (O6-BG) is a nonreversible inhibitor of MGMT, it promotes the cytotoxicity of alkylating chemotherapy. The authors have developed a hybrid-structured nanofibrous membrane (HSNM) that sequentially delivers high concentrations of O6-BG, BiCNU, and TMZ in an attempt to provide an alternative to the current therapeutic options for MGs. METHODS: The HSNMs were implanted onto the cerebral surface of pathogen-free rats following surgical craniectomy, while the in vivo release behaviors of O6-BG, TMZ, and BiCNU from the HSNMs were explored. Subsequently, the HSNMs were surgically implanted onto the brain surface of two types of tumor-bearing rats. The survival rate, tumor volume, malignancy of tumor, and apoptotic cell death were evaluated and compared with other treatment regimens. RESULTS: The biodegradable HSNMs sequentially and sustainably delivered high concentrations of O6-BG, BiCNU, and TMZ for more than 14 weeks. The tumor-bearing rats treated with HSNMs demonstrated therapeutic advantages in terms of retarded and restricted tumor growth, prolonged survival time, and attenuated malignancy. CONCLUSION: The results demonstrated that O6-BG potentiates the effects of interstitially transported BiCNU and TMZ. Therefore, O6-BG may be required for alkylating agents to offer maximum therapeutic benefits for the treatment of MGMT-expressing tumors. In addition, the HSNM-supported chemoprotective gene therapy enhanced chemotherapy tolerance and efficacy. It can, therefore, potentially provide an improved therapeutic alternative for MGs.

13.
J Biomed Sci ; 26(1): 77, 2019 Oct 19.
Article in English | MEDLINE | ID: mdl-31629402

ABSTRACT

BACKGROUND: Intratumor subsets with tumor-initiating features in glioblastoma are likely to survive treatment. Our goal is to identify the key factor in the process by which cells develop temozolomide (TMZ) resistance. METHODS: Resistant cell lines derived from U87MG and A172 were established through long-term co-incubation of TMZ. Primary tumors obtained from patients were maintained as patient-derived xenograft for studies of tumor-initating cell (TIC) features. The cell manifestations were assessed in the gene modulated cells for relevance to drug resistance. RESULTS: Among the mitochondria-related genes in the gene expression databases, superoxide dismutase 2 (SOD2) was a significant factor in resistance and patient survival. SOD2 in the resistant cells functionally determined the cell fate by limiting TMZ-stimulated superoxide reaction and cleavage of caspase-3. Genetic inhibition of the protein led to retrieval of drug effect in mouse study. SOD2 was also associated with the TIC features, which enriched in the resistant cells. The CD133+ specific subsets in the resistant cells exhibited superior superoxide regulation and the SOD2-related caspase-3 reaction. Experiments applying SOD2 modulation showed a positive correlation between the TIC features and the protein expression. Finally, co-treatment with TMZ and the SOD inhibitor sodium diethyldithiocarbamate trihydrate in xenograft mouse models with the TMZ-resistant primary tumor resulted in lower tumor proliferation, longer survival, and less CD133, Bmi-1, and SOD2 expression. CONCLUSION: SOD2 plays crucial roles in the tumor-initiating features that are related to TMZ resistance. Inhibition of the protein is a potential therapeutic strategy that can be used to enhance the effects of chemotherapy.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Superoxide Dismutase/administration & dosage , Temozolomide/pharmacology , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Heterografts/physiopathology , Humans , Mice , Neoplastic Stem Cells/physiology
14.
World Neurosurg ; 132: 208-210, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31505278

ABSTRACT

A 76-year-old man presented with progressive dementia, gait disturbance, and urinary incontinence for 1 year. Computed tomography scan revealed nonobstructive hydrocephalus, but abnormal papillary structures at the ventricular wall were noted. Before cerebrospinal fluid (CSF) diversion surgery for hydrocephalus, we performed magnetic resonance angiography and magnetic resonance venography, which revealed multiple engorged vessels over the ventricular wall and bilateral hemispheres. Digital subtraction angiography revealed 2 dural arteriovenous fistulas (DAVFs) at the left transverse-sigmoid sinus and superior sagittal sinus. Signs of angioarchitecture characteristic of cerebral venous hypertension (CVH) were noted, including cortical vein regurgitation and severe pseudophlebitic pattern. DAVFs with CVH might be a factor contributing to acquired hydrocephalus. DAVFs should be considered when patients with hydrocephalus exhibit abnormal papillary structures at the ventricular wall. Performing CSF diversion surgery for hydrocephalus before downgrading or curing such aggressive DAVFs may lead to major complications.


Subject(s)
Central Nervous System Vascular Malformations/complications , Cerebral Ventricles/pathology , Hydrocephalus/etiology , Aged , Angiography, Digital Subtraction , Central Nervous System Vascular Malformations/pathology , Central Nervous System Vascular Malformations/surgery , Cerebral Ventricles/surgery , Humans , Hydrocephalus/pathology , Hydrocephalus/surgery , Hypertension/etiology , Magnetic Resonance Imaging , Male , Neurosurgical Procedures/methods , Tomography, X-Ray Computed , Treatment Outcome
15.
J Neurosurg ; 132(5): 1566-1573, 2019 Apr 26.
Article in English | MEDLINE | ID: mdl-31026826

ABSTRACT

OBJECTIVE: A high level of vascular endothelial growth factor (VEGF) has been implicated in brain arteriovenous malformation (bAVM) bleeding and rupture. However, direct evidence is missing. In this study the authors used a mouse bAVM model to test the hypothesis that elevation of focal VEGF levels in bAVMs exacerbates the severity of bAVM hemorrhage. METHODS: Brain AVMs were induced in adult mice in which activin receptor-like kinase 1 (Alk1, a gene that causes AVM) gene exons 4-6 were floxed by intrabasal ganglia injection of an adenoviral vector expressing Cre recombinase to induce Alk1 mutation and an adeno-associated viral vector expressing human VEGF (AAV-VEGF) to induce angiogenesis. Two doses of AAV-VEGF (5 × 109 [high] or 2 × 109 [low]) viral genomes were used. In addition, the common carotid artery and external jugular vein were anastomosed in a group of mice treated with low-dose AAV-VEGF 6 weeks after the model induction to induce cerebral venous hypertension (VH), because VH increases the VEGF level in the brain. Brain samples were collected 8 weeks after the model induction. Hemorrhages in the bAVM lesions were quantified on brain sections stained with Prussian blue, which detects iron deposition. VEGF levels were quantified in bAVM tissue by enzyme-linked immunosorbent assay. RESULTS: Compared to mice injected with a low dose of AAV-VEGF, the mice injected with a high dose had higher levels of VEGF (p = 0.003) and larger Prussian blue-positive areas in the bAVM lesion at 8 or 9 weeks after model induction (p = 0.002). VH increased bAVM hemorrhage in the low-dose AAV-VEGF group. The overall mortality in the high-dose AAV-VEGF group was 26.7%, whereas no mouse died in the low-dose AAV-VEGF group without VH. In contrast, VH caused a mortality of 50% in the low-dose AAV-VEGF group. CONCLUSIONS: Using mouse bAVM models, the authors provided direct evidence that elevation of the VEGF level increases bAVM hemorrhage and mouse mortality.

16.
Phytomedicine ; 49: 41-51, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30217261

ABSTRACT

BACKGROUND: Temozolomide (TMZ) is a first-line chemotherapeutic drug for malignant gliomas. Nonetheless, TMZ-induced side effects and drug resistance remain challenges. Our previous study showed the suppressive effects of honokiol on growth of gliomas. PURPOSE: This study was further aimed to evaluate if honokiol could enhance TMZ-induced insults toward malignant glioma cells and its possible mechanisms. METHODS: Human U87 MG glioma cells were exposed to TMZ, honokiol, and a combination of TMZ and honokiol. Cell survival, apoptosis, necrosis, and proliferation were successively assayed. Fluorometric substrate assays were conducted to determine activities of caspase-3, -6, -8, and -9. Levels of Fas ligand, Bax, and cytochrome c were immunodetected. Translocation of Bax to mitochondria were examined using confocal microscopy. Mitochondrial function was evaluated by assaying the mitochondrial membrane potential (MMP), reactive oxygen species (ROS), and complex I enzyme activity. Caspase-6 activity was suppressed using specific peptide inhibitors. The honokiol-induced effects were further confirmed using human U373 MG and murine GL261 cells. RESULTS: Exposure of human U87 MG glioma cells to honokiol significantly increased TMZ-induced DNA fragmentation and cell apoptosis. Interestingly, honokiol enhanced intrinsic caspase-9 activity without affecting extrinsic Fas ligand levels and caspase-8 activity. Sequentially, TMZ-induced changes in Bax translocation, the MMP, mitochondrial complex I enzyme activity, intracellular ROS levels, and cytochrome c release were enhanced by honokiol. Consequently, honokiol amplified TMZ-induced activation of caspases-3 and -6 in human U87 MG cells. Fascinatingly, suppressing caspase-6 activity concurrently decreased honokiol-induced DNA fragmentation and cell apoptosis. The honokiol-involved improvement in TMZ-induced intrinsic apoptosis was also confirmed in human U373 MG and murine GL261 glioma cells. CONCLUSIONS: This study showed that honokiol can enhance TMZ-induced apoptotic insults to glioma cells via an intrinsic mitochondrion-dependent mechanism. Our results suggest the therapeutic potential of honokiol to attenuate TMZ-induced side effects.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Dacarbazine/analogs & derivatives , Drugs, Chinese Herbal/pharmacology , Glioma/pathology , Lignans/pharmacology , Mitochondria/physiology , Animals , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , DNA Fragmentation , Dacarbazine/pharmacology , Fas Ligand Protein/metabolism , Glioma/drug therapy , Humans , Membrane Potential, Mitochondrial , Mice , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Temozolomide
18.
Mol Oncol ; 12(5): 756-771, 2018 05.
Article in English | MEDLINE | ID: mdl-29603594

ABSTRACT

Glioblastoma multiforme (GBM) is a highly malignant type of brain tumor found in humans. GBM cells reproduce quickly, and the median survival time for patients after therapy is approximately 1 year with a high relapse rate. Current therapies and diagnostic tools for GBM are limited; therefore, we searched for a more favorable therapeutic target or marker protein for both therapy and diagnosis. We used mass spectrometry (MS) analysis to identify GBM-associated marker proteins from human plasma and GBM cell cultures. Additional plasma and 52 brain tissues obtained from patients with gliomas were used to validate the association rate of serum amyloid A1 (SAA1) in different grades of gliomas and its distribution in tumors. Microarray database analysis further validated the coefficient of SAA1 levels in gliomas. The cellular mechanisms of SAA1 in GBM proliferation and infiltration were investigated in vitro. We analyzed the correlation between SAA1 and patients' medication requirement to demonstrate the clinical effects of SAA1 in GBM. SAA1 was identified from MS analysis, and its level was revealed to be correlated with the disease grade, clinical severity, and survival rate of patients with gliomas. In vitro cultures, including GBM cells and normal astrocytes, revealed that SAA1 promotes cell migration and invasion through integrin αVß3 to activate the Erk signaling pathway. Magnetic resonance imaging and tumor region-specific microarray analysis identified a correlation between SAA1 and GBM cell infiltration in patients. In summary, our results demonstrate that SAA1 in combination with integrin αV and ß3 can serve as an indicator of high glioblastoma risk. We also identified the cellular mechanisms of SAA1 contributing to GBM progression, which can serve as the basis for future GBM therapy.


Subject(s)
Cell Movement , Disease Progression , Glioblastoma/metabolism , Glioblastoma/pathology , Integrin alphaVbeta3/metabolism , Serum Amyloid A Protein/metabolism , Astrocytes/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Female , Glioblastoma/blood , Glioblastoma/diagnosis , Humans , Male , Middle Aged , Neoplasm Invasiveness , Survival Analysis
19.
Artif Cells Nanomed Biotechnol ; 46(sup2): 515-526, 2018.
Article in English | MEDLINE | ID: mdl-29658349

ABSTRACT

Chemotherapy is ineffective for treating malignant glioma (MG) because of the low therapeutic levels of pharmaceuticals in tumour tissues and the well-known tumour resistance. The resistance to alkylators is modulated by the DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT). O6-benzylguanine (O6-BG) can irreversibly inactivate AGT by competing with O6-methylguanine and has been confirmed to increase the therapeutic activity of alkylators. We developed hybrid-structured poly[(d,l)-lactide-co-glycolide] nanofibrous membranes (HSNMs) that enable the sequential and sustained release of O6-BG and two alkylators (carmustine and temozolomide [TMZ]). HSNMs were surgically instilled into the cerebral cavity of pathogen-free rats and F98 glioma-bearing rats. The release behaviours of loaded drugs were quantified by using high-performance liquid chromatography. The treatment results were compared with the rats treated with intraperitoneal injection of O6-BG combined with surgical implantation of carmustine wafer and oral TMZ. The HSNMs revealed a sequential drug release behaviour with the elution of high drug concentrations of O6-BG in the early phase, followed by high levels of two alkylators. All drug concentrations remained high for over 14 weeks. Tumour growth was slower and the mean survival time was significantly prolonged in the HSNM-treated group. Biodegradable HSNMs can enhance therapeutic efficacy and prevent toxic systemic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Genetic Therapy , Glioma/pathology , Glioma/therapy , Nanofibers/chemistry , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Liberation , Glioma/drug therapy , Glioma/genetics , Magnetic Resonance Imaging , Male , Rats , Survival Analysis , Tumor Burden/drug effects
20.
Oncol Lett ; 15(1): 91-98, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29375706

ABSTRACT

Radiosurgery serves an important function in the treatment of patients with intraocular tumors and preserves visual function via organ conservation. Therefore, it is important to ensure the safety and precision of GK-SRS as a primary treatment for intraocular tumors. The present case study described a 57-year-old female with uveal melanoma treated with GK-SRS. Retrobulbar anesthesia following fixation of the treated eye, via the suture of two of the extraocular muscles to the stereotactic frame, was performed to immobilize the eye during treatment. Computed tomography (CT) scans were performed following eye fixation, immediately prior to and following GK-SRS, to validate the accuracy of the tumor localization. The eye movement analysis revealed that the gravity center point deviations of the tumor and lens during treatment were <0.110 mm. At least 95% of the tumor volume was covered by the prescription dose according to three sets of CT images. The patient underwent a trans pars plana vitrectomy owing to a right eye vitreous hemorrhage. A 37-month follow-up assessment revealed tumor shrinkage, and the disappearance of the serous retinal detachments was noted on the basis of ophthalmoscopy and orbital magnetic resonance imaging. No major complications developed during the follow-up period. Using our treatment protocol, GK-SRS is a non-invasive procedure which is used as a brief single fraction treatment for intraocular tumor. The eye fixation method used in the present study has high accuracy.

SELECTION OF CITATIONS
SEARCH DETAIL
...