Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Neurooncol ; 161(2): 193-202, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35612696

RESUMEN

OBJECTIVE: To evaluate the clinicopathological characteristics, radiology, and long-term outcomes of microcystic meningiomas (MM) and compare it with other subtypes of meningiomas managed at a single neurosurgical center. METHODS: A total of 87 consecutive patients who underwent surgical resection and were diagnosed as MM between 2005 and 2016 were enrolled for analysis. Clinicopathological, radiology, and prognostic information was collected and analyzed. Progression free survival (PFS) was compared with 659 patients with other subtypes of WHO grade 1 meningiomas and 167 patients with atypical meningiomas treated during the same period. RESULTS: Fifty six females and 31 males with MM were analyzed. Peri-tumor brain edema was frequent on T2 WI (85%).12 patients (13.8%) experienced tumor progression during the mean follow-up of 101.66 ± 40.92 months. The median PFS was unavailable, and the 5, 10, and 15 year progression-free rates were 96.9%, 84.0%, and 73.9%, respectively. Univariate COX analysis demonstrated skull base location and higher Ki-67 index as significant negative prognostic factors for PFS (P < 0.05); multivariate analysis identified tumor location and Ki-67 index as independent factors (P < 0.01), as well. Of note, the PFS of MM was worse than other WHO grade 1 subtypes (P < 0.001), but better than atypical meningiomas (P < 0.001), and the PFS differences were retained even when the analysis was limited to the patients receiving GTR (P < 0.05). CONCLUSION: The PFS of MM was worse than other WHO grade 1 subtypes and better than atypical meningiomas. Skull base location and higher Ki-67 index were independent negative prognostic factors in MM.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Masculino , Femenino , Humanos , Meningioma/diagnóstico por imagen , Meningioma/cirugía , Neoplasias Meníngeas/cirugía , Neoplasias Meníngeas/diagnóstico , Antígeno Ki-67 , Pronóstico , Organización Mundial de la Salud
2.
Cell Mol Neurobiol ; 41(8): 1651-1663, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32770297

RESUMEN

Concussion is a widely recognized environmental risk factor for neurodegenerative diseases, including Parkinson's disease (PD). Small-vessel disease of the brain has been reported to contribute to neurodegenerative diseases. In this study, we observed BBB disruption in wild-type (WT) mice, but not in matrix metalloproteinase 9 (MMP-9) knockout mice, subjected to single severe traumatic brain injury (ssTBI). Furthermore, treating ssTBI mice with the MMP-9 inhibitor GM6001 effectively maintained BBB integrity, promoted the elimination of damaged mitochondria via mitophagy, and then prevented neuronal death and progressive neurodegeneration. However, we did not observe this neuroprotective effect of MMP-9 inhibition in beclin-1-/+ mice. Collectively, these findings revealed that concussion led to BBB disruption via MMP-9, and that GM6001 prevented the development of PD via the autophagy pathway.


Asunto(s)
Autofagia/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Dipéptidos/uso terapéutico , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Trastornos Parkinsonianos/tratamiento farmacológico , Animales , Autofagia/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/enzimología , Lesiones Traumáticas del Encéfalo/patología , Dipéptidos/farmacología , Femenino , Masculino , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Parkinsonianos/enzimología , Trastornos Parkinsonianos/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Índices de Gravedad del Trauma
3.
Pharmacol Res ; 174: 105933, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34634471

RESUMEN

Ischemic stroke poses a significant health risk due to its high rate of disability and mortality. To address this problem, several therapeutic approaches have been proposed, including interruption targeting programmed cell death (PCD). Ferroptosis is a newly defined PCD characterized by iron-dependent accumulation of lipid peroxidation, and is becoming a promising target for treating numerous diseases. To explore the underlying mechanisms of the initiation and execution of ferroptosis in ischemic stroke, we established stroke models in vivo and in vitro simulating ischemia/reperfusion (I/R) neuronal injury. Different from previous reports on stroke, we tested ferroptosis by measuring the levels of core proteins, such as ACSL4, 15-LOX2, Ferritin and GPX4. In addition, I/R injury induces excessive degradation of ferritin via the autophagy pathway and subsequent increase of free iron in neurons. This phenomenon has recently been termed ferritinophagy and reported to be regulated by nuclear receptor coactivator 4 (NCOA4) in some cell lines. Increased NCOA4 in cytoplasm was detected in our study and then silenced by shRNA to investigate its function. Both in vivo and in vitro, NCOA4 deletion notably abrogated ferritinophagy caused by I/R injury and thus inhibited ferroptosis. Furthermore, we found that NCOA4 was upregulated by ubiquitin specific peptidase 14 (USP14) via a deubiquitination process in damaged neurons, and we found evidence of pharmacological inhibition of USP14 effectively reducing NCOA4 levels to protect neurons from ferritinophagy-mediated ferroptosis. These findings suggest a novel and effective target for treating ischemic stroke.


Asunto(s)
Ferroptosis , Infarto de la Arteria Cerebral Media , Accidente Cerebrovascular Isquémico , Coactivadores de Receptor Nuclear , Daño por Reperfusión , Animales , Encéfalo/metabolismo , Células Cultivadas , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Accidente Cerebrovascular Isquémico/genética , Accidente Cerebrovascular Isquémico/metabolismo , Peroxidación de Lípido , Masculino , Malondialdehído/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Coactivadores de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/metabolismo , Pirroles/farmacología , Pirrolidinas/farmacología , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo
4.
J Neurosci ; 39(10): 1930-1943, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30626699

RESUMEN

Mitochondrial energy production is essential for normal brain function. Traumatic brain injury (TBI) increases brain energy demands, results in the activation of mitochondrial respiration, associated with enhanced generation of reactive oxygen species. This chain of events triggers neuronal apoptosis via oxidation of a mitochondria-specific phospholipid, cardiolipin (CL). One pathway through which cells can avoid apoptosis is via elimination of damaged mitochondria by mitophagy. Previously, we showed that externalization of CL to the mitochondrial surface acts as an elimination signal in cells. Whether CL-mediated mitophagy occurs in vivo or its significance in the disease processes are not known. In this study, we showed that TBI leads to increased mitophagy in the human brain, which was also detected using TBI models in male rats. Knockdown of CL synthase, responsible for de novo synthesis of CL, or phospholipid scramblase-3, responsible for CL translocation to the outer mitochondrial membrane, significantly decreased TBI-induced mitophagy. Inhibition of mitochondrial clearance by 3-methyladenine, mdivi-1, or phospholipid scramblase-3 knockdown after TBI led to a worse outcome, suggesting that mitophagy is beneficial. Together, our findings indicate that TBI-induced mitophagy is an endogenous neuroprotective process that is directed by CL, which marks damaged mitochondria for elimination, thereby limiting neuronal death and behavioral deficits.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) increases energy demands leading to activation of mitochondrial respiration associated with enhanced generation of reactive oxygen species and resultant damage to mitochondria. We demonstrate that the complete elimination of irreparably damaged organelles via mitophagy is activated as an early response to TBI. This response includes translocation of mitochondria phospholipid cardiolipin from the inner membrane to the outer membrane where externalized cardiolipin mediates targeted protein light chain 3-mediated autophagy of damaged mitochondria. Our data on targeting phospholipid scramblase and cardiolipin synthase in genetically manipulated cells and animals strongly support the essential role of cardiolipin externalization mechanisms in the endogenous reparative plasticity of injured brain cells. Furthermore, successful execution and completion of mitophagy is beneficial in the context of preservation of cognitive functions after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Encéfalo/metabolismo , Cardiolipinas/metabolismo , Mitofagia/fisiología , Neuronas/metabolismo , Animales , Apoptosis/fisiología , Encéfalo/ultraestructura , Lesiones Traumáticas del Encéfalo/patología , Humanos , Masculino , Membranas Mitocondriales/metabolismo , Neuronas/ultraestructura , Ratas Sprague-Dawley , Transducción de Señal
5.
J Cell Mol Med ; 24(15): 8466-8479, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32585748

RESUMEN

Microglial cells are key component of central nervous system (CNS) and mediate the immune response of the brain under physiological or pathological conditions. It tends to activate into a pro-inflammatory M1 phenotype after traumatic brain injury (TBI) and promote secondary brain damage. Recently, necroptosis was found to promote microglial activation and neuroinflammation after TBI. However, the mechanism and specific interventions of microglial necroptosis after TBI remain poorly investigated. Here, we reported that overexpress the charged multivesicular body protein 4b (CHMP4B) which is a core member of the endosomal sorting required for transport complex III (ESCRT-III) significantly decreased the level of necroptosis in microglia, improved neurological function recovery and protected against cell death after TBI. Further investigation showed that forkhead transcription factor O1 (FOXO1) was a crucial transcription factor that increased CHMP4B transcription by binding to the promoter region, thereby inhibiting necroptosis in microglia. Collectively, our findings demonstrated that CHMP4B relieved microglial necroptosis and neuroinflammation after TBI, and promote the recovery of nerve function. FOXO1 is an important factor in promoting CHMP4B expression. This study provides the novel viewpoint for TBI prevention and treatment.


Asunto(s)
Lesiones Traumáticas del Encéfalo/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Microglía/patología , Necroptosis/genética , Regulación hacia Arriba/genética , Adulto , Anciano , Animales , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Línea Celular , Femenino , Proteína Forkhead Box O1/genética , Regulación de la Expresión Génica/genética , Humanos , Inflamación/genética , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , Adulto Joven
6.
J Cell Mol Med ; 24(2): 1474-1487, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31856394

RESUMEN

Tumour invasion is closely related to the prognosis and recurrence of glioblastoma multiforme and partially attributes to epithelial-mesenchymal transition. Long intergenic non-coding RNA 00511 (LINC00511) plays a pivotal role in tumour; however, the role of LINC00511 in GBM, especially in the epigenetic molecular regulation mechanism of EMT, is still unclear. Here, we found that LINC00511 was up-regulated in GBM tissues and relatively high LINC00511 expression predicted poorer prognosis. Moreover, ectopic LINC00511 enhanced GBM cells proliferation, EMT, migration and invasion, whereas LINC00511 knockdown had the opposite effects. Mechanistically, we confirmed that ZEB1 acted as a transcription factor for LINC00511 in GBM cells. Subsequently, we found that LINC00511 served as a competing endogenous RNA that sponged miR-524-5p to indirectly regulate YB1, whereas, up-regulated YB1 promoted ZEB1 expression, which inversely facilitated LINC00511 expression. Finally, orthotopic xenograft models were performed to further demonstrate the LINC00511 on GBM tumorigenesis. This study demonstrates that a LINC00511/miR-524-5p/YB1/ZEB1 positive feedback loop provides potential therapeutic targets for GBM progression.


Asunto(s)
Carcinogénesis/genética , Transición Epitelial-Mesenquimal/genética , Retroalimentación Fisiológica , Glioblastoma/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Secuencia de Bases , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Carcinogénesis/patología , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Células HEK293 , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica , Pronóstico , ARN Largo no Codificante/genética , Transcripción Genética , Regulación hacia Arriba/genética
7.
J Cell Mol Med ; 23(10): 6907-6918, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31430050

RESUMEN

Glioblastoma (GBM) is the most universal type of primary brain malignant tumour, and the prognosis of patients with GBM is poor. S100A11 plays an essential role in tumour. However, the role and molecular mechanism of S100A11 in GBM are not clear. Here, we found that S100A11 was up-regulated in GBM tissues and higher S100A11 expression indicated poor prognosis of GBM patients. Overexpression of S100A11 promoted GBM cell growth, epithelial-mesenchymal transition (EMT), migration, invasion and generation of glioma stem cells (GSCs), whereas its knockdown inhibited these activities. More importantly, S100A11 interacted with ANXA2 and regulated NF-κB signalling pathway through decreasing ubiquitination and degradation of ANXA2. Additionally, NF-κB regulated S100A11 at transcriptional level as a positive feedback. We also demonstrated the S100A11 on tumour growth in GBM using an orthotopic tumour xenografting. These data demonstrate that S100A11/ANXA2/NF-κB positive feedback loop in GBM cells that promote the progression of GBM.


Asunto(s)
Anexina A2/metabolismo , Neoplasias Encefálicas/genética , Retroalimentación Fisiológica , Glioblastoma/genética , FN-kappa B/metabolismo , Oncogenes , Proteínas S100/metabolismo , Animales , Neoplasias Encefálicas/patología , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Pronóstico , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteolisis , Transducción de Señal , Esferoides Celulares/patología , Transcripción Genética , Ubiquitinación , Regulación hacia Arriba/genética
8.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1663-1674, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29466698

RESUMEN

Acute lung injury (ALI) is one of several complications in patients with traumatic brain injury (TBI). Autophagy is a primary homeostatic process that promotes cell survival under stress. Accumulating evidence implicates autophagy in the pathogenesis of ALI under various conditions. However, the role of autophagy in TBI-induced ALI remains unknown. The aim of this study was to adjust autophagy with pharmacological agents to determine its functional significance in TBI-induced ALI. Rats were preconditioned with autophagy promoter rapamycin or inhibitor 3-methyladenine before they were challenged with TBI. Extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor U0126, mechanistic target of rapamycin (mTOR) inhibitor rapamycin, and signal transducer and activator of transcription 3 (Stat3) inhibitor S31-201 were used to test the role of ERK1/2/mTOR/Stat3 signaling pathway in regulating autophagy. Autophagy is activated in lung tissues after TBI. Enhancement of autophagy suppressed apoptosis, inflammation and oxidative stress in lung tissues, which were activated after TBI, whereas inhibition of autophagy aggravated these critical pathological changes. Autophagy also improved TBI-induced impairment in pulmonary barrier function, oxygenation function and static compliance. Furthermore, TBI-induced autophagy was mediated by ERK1/2/mTOR/Stat3 pathway, which may serve to reduce ALI and improve pulmonary barrier function, oxygenation function and static compliance. These findings are important for the prevention and treatment of TBI-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Autofagia , Lesiones Traumáticas del Encéfalo/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor de Transcripción STAT3/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Apoptosis , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Pulmón/metabolismo , Pulmón/patología , Masculino , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(9 Pt B): 2957-2971, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29894755

RESUMEN

Phospholipase A2 is a known aggravator of inflammation and deteriorates neurological outcomes after traumatic brain injury (TBI), however the exact inflammatory mechanisms remain unknown. This study investigated the role of bradykinin and its receptor, which are known initial mediators within inflammation activation, as well as the mechanisms of the cytosolic phospholipase A2 (cPLA2)-related inflammatory responses after TBI. We found that cPLA2 and bradykinin B2 receptor were upregulated after a TBI. Rats treated with the bradykinin B2 receptor inhibitor LF 16-0687 exhibited significantly less cPLA2 expression and related inflammatory responses in the brain cortex after sustaining a controlled cortical impact (CCI) injury. Both the cPLA2 inhibitor and the LF16-0687 improved CCI rat outcomes by decreasing neuron death and reducing brain edema. The following TBI model utilized both primary astrocytes and primary neurons in order to gain further understanding of the inflammation mechanisms of the B2 bradykinin receptor and the cPLA2 in the central nervous system. There was a stronger reaction from the astrocytes as well as a protective effect of LF16-0687 after the stretch injury and bradykinin treatment. The protein kinase C pathway was thought to be involved in the B2 bradykinin receptor as well as the cPLA2-related inflammatory responses. Rottlerin, a Protein Kinase C (PKC) δ inhibitor, decreased the activity of the cPLA2 activity post-injury, and LF16-0687 suppressed both the PKC pathway and the cPLA2 activity within the astrocytes. These results indicated that the bradykinin B2 receptor-mediated pathway is involved in the cPLA2-related inflammatory response from the PKC pathway.


Asunto(s)
Bradiquinina/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Inflamación/patología , Fosfolipasas A2 Citosólicas/metabolismo , Receptor de Bradiquinina B2/metabolismo , Acetofenonas/farmacología , Adulto , Anciano , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/patología , Benzopiranos/farmacología , Bradiquinina/administración & dosificación , Bradiquinina/sangre , Bradiquinina/líquido cefalorraquídeo , Antagonistas del Receptor de Bradiquinina B2/farmacología , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/sangre , Lesiones Traumáticas del Encéfalo/líquido cefalorraquídeo , Lesiones Traumáticas del Encéfalo/etiología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Epilepsia/líquido cefalorraquídeo , Epilepsia/patología , Femenino , Humanos , Inflamación/sangre , Inflamación/líquido cefalorraquídeo , Inflamación/etiología , Masculino , Persona de Mediana Edad , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba , Adulto Joven
10.
Cell Mol Neurobiol ; 37(8): 1359-1371, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28124208

RESUMEN

Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. The extraordinary invasion of human GBM into adjacent normal brain tissues contributes to treatment failure. However, the mechanisms that control this process remain poorly understood. Increasing evidence has demonstrated that microRNAs are strongly implicated in the migration and invasion of GBM. In this study, we found that microRNA-98 (miR-98) was markedly downregulated in human glioma tissues and cell lines. Functional experiments indicated that restored expression of miR-98 attenuated glioma cell invasion and migration, whereas depletion of miR-98 promoted glioma cell invasion and migration. Subsequent investigation showed that pre-B-cell leukemia homeobox 3 (PBX3), an important transcription factor that controls tumor invasion, was a direct and functional target of miR-98 in GBM cells. Consistently, an orthotopic mouse model also demonstrated the suppressive effects of miR-98 overexpression on tumor invasion and PBX3 expression. Silencing of PBX3 using small interfering RNA inhibited the migratory and invasive capacities of glioma cells, whereas reintroduction of PBX3 into glioma cells reversed the anti-invasive function of miR-98. Furthermore, depletion of PBX3 phenocopied the effects of miR-98 overexpression in vivo. Finally, quantitative real-time polymerase chain reaction results showed that miR-98 was negatively correlated with PBX3 expression in 24 glioma tissues. Thus, we propose that PBX3 modulation by miR-98 has an important role in regulating GBM invasion and may serve as therapeutic target for GBM.


Asunto(s)
Neoplasias Encefálicas/genética , Movimiento Celular/fisiología , Marcación de Gen/métodos , Glioma/genética , Proteínas de Homeodominio/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Proteínas de Homeodominio/biosíntesis , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/biosíntesis , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas Proto-Oncogénicas/biosíntesis
11.
J Neurooncol ; 135(3): 453-463, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28856521

RESUMEN

Among primary brain tumors, gliomas are the most common and most aggressive, with a poor prognosis and limited treatment options. Thus, it is essential to determine the mechanisms involved in glioma development to develop effective therapies for glioma patients. Pre-B-cell leukemia homeobox 3 (PBX3), a critical member of the PBX family, is frequently overexpressed in multiple human malignancies. However, the expression patterns and biological functions, as well as the involved molecular functions of PBX3 in human gliomas remain largely unknown. In this study, we demonstrate that PBX3 expression is increased in both human glioma tissues and cell lines compared with their normal counterparts. These results suggested that PBX3 might be involved in glioma progression. Thus, the role of PBX3 in glioma cell proliferation was investigated using genetic knockdown and overexpression methods. The results showed that PBX3 knockdown inhibited glioma cell proliferation and induced apoptosis, while PBX3 overexpression significantly promoted glioma cell proliferation. Mechanistically, we found that PBX3 promoted cell proliferation by modulating cell cycle progression. A xenograft LN229 model was used to confirm that PBX3 depletion decreased tumor growth in vivo. In summary, our findings reveal that PBX3 may be a potential therapeutic target in gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proliferación Celular/fisiología , Glioma/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Apoptosis/fisiología , Neoplasias Encefálicas/patología , Ciclo Celular/fisiología , Línea Celular Tumoral , Minería de Datos , Técnicas de Silenciamiento del Gen , Glioma/patología , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas/genética , Regulación hacia Arriba
12.
Redox Biol ; 72: 103137, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38642502

RESUMEN

The oncogene Aurora kinase A (AURKA) has been implicated in various tumor, yet its role in meningioma remains unexplored. Recent studies have suggested a potential link between AURKA and ferroptosis, although the underlying mechanisms are unclear. This study presented evidence of AURKA upregulation in high grade meningioma and its ability to enhance malignant characteristics. We identified AURKA as a suppressor of erastin-induced ferroptosis in meningioma. Mechanistically, AURKA directly interacted with and phosphorylated kelch-like ECH-associated protein 1 (KEAP1), thereby activating nuclear factor erythroid 2 related factor 2 (NFE2L2/NRF2) and target genes transcription. Additionally, forkhead box protein M1 (FOXM1) facilitated the transcription of AURKA. Suppression of AURKA, in conjunction with erastin, yields significant enhancements in the prognosis of a murine model of meningioma. Our study elucidates an unidentified mechanism by which AURKA governs ferroptosis, and strongly suggests that the combination of AURKA inhibition and ferroptosis-inducing agents could potentially provide therapeutic benefits for meningioma treatment.


Asunto(s)
Aurora Quinasa A , Ferroptosis , Proteína Forkhead Box M1 , Meningioma , Factor 2 Relacionado con NF-E2 , Piperazinas , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Proteína Forkhead Box M1/metabolismo , Proteína Forkhead Box M1/genética , Aurora Quinasa A/metabolismo , Aurora Quinasa A/genética , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Animales , Ratones , Meningioma/metabolismo , Meningioma/genética , Meningioma/patología , Piperazinas/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/tratamiento farmacológico , Resistencia a Antineoplásicos/genética
14.
Front Oncol ; 13: 1133861, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36816938

RESUMEN

Background: The treatment for giant pituitary adenomas (GPAs, maximal diameter >4 cm) remains challenging, with remarkable mortality and morbidity, and there is no consensus on the optimal surgical approach. Gross total resection (GTR) for GPAs is difficult to achieve through a single transsphenoidal or transcranial approach. Any residual tumor is at risk for postoperative apoplexy. In this study, we propose a new surgical technique for resecting the GPAs in a sing-stage transcranial surgery. Methods: A retrospective review of 4 patients with complicated GPAs, who had been treated via an endoscopic transcranial transdiaphragmatic approach in a single-stage surgery after routine transcranial resection, was performed. The following data was analyzed: clinical characteristics, preoperative imaging studies, resection rate, perioperative morbidity and mortality, as well as postoperative outcomes. Results: All patients had nonfunctioning GPAs and preoperative visual disturbances. In three patients, GTR was achieved, and in one patient, near-total resection (90%-100% of the tumor) was achieved. Three patients attained improved postoperative visual function, while one patient's vision remained unchanged. One patient suffered a deficiency in adrenocorticotropic hormone along with thyroid-stimulating hormone, and one patient developed diabetes insipidus. Notably, none of the patients suffered cerebrospinal fluid leakage. However, one patient developed an epidural hematoma and underwent decompressive craniectomy. Conclusions: The endoscopic transcranial transdiaphragmatic approach in a single-stage surgery can be efficiently and safely performed for maximal excision of GPAs with extensive suprasellar extension. Furthermore, relative to the conventional combined or staged approaches, this innovative surgical strategy provides neurosurgeons with a clear operative field with reduced invasiveness.

15.
Acad Radiol ; 30(11): 2469-2476, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36697269

RESUMEN

RATIONALE AND OBJECTIVES: The measurement of the time since stroke onset (TSS) is crucial for decision-making in the treatment of acute ischemic stroke (AIS). This study assessed the utility of computed tomography angiography (CTA) radiomics features (RFs) to estimate TSS. MATERIALS AND METHODS: A total of 221 patients with AIS were enrolled in this retrospective study and were divided into a training group (n = 154) and a test group (n = 67). Thrombi in CTA images were manually outlined using ITK-SNAP. Images were aligned, normalized, and pre-processed to extract RFs. The TSS was calculated as the time from stroke onset to CTA completion. The patients were classified into two groups according to estimated TSS: ≤4.5 and >4.5 hours. A total of 944 RFs were extracted from CTA images. Clinical factors associated with TSS were identified using multivariate logistic regression, and a combined model (clinical data and RFs) was constructed. The predictive value of the models was assessed by the area under the receiver operating characteristic curve (AUC). The performance of the models was compared using the DeLong test, and clinical utility was evaluated by decision curve analysis. RESULTS: The AUC of the radiomics model was 0.803 (95% confidence interval [CI]: 0.733-0.873) and 0.803 (95% CI: 0.698-0.908) in the training and test cohorts, respectively. The AUC of the combined model (containing data on age, diabetes, and atrial fibrillation) in the training and test sets was 0.813 (95% CI: 0.750-0.889) and 0.803 (95% CI: 0.699-0.907), respectively. The DeLong test showed no significant difference between the radiomics and combined models. Decision curve analysis showed that both models had clinical utility. CONCLUSION: CTA-based thrombus radiomics can estimate TSS in patients with AIS. The addition of clinical data to the model does not improve predictive performance.

16.
J Mol Cell Biol ; 2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37791390

RESUMEN

Meningioma is one of the most common primary neoplasms in the central nervous system, whereas there is still no specific molecularly targeted therapy that has been approved for the clinical treatment of aggressive meningiomas. There is therefore an urgent demand to decrypt the biological and molecular landscape of malignant meningioma. Here, through the in-silica prescreening and 10-year follow-up of 445 meningioma patients, we uncovered that CBX7 is progressively decreased with malignancy grade and neoplasia stage in meningioma and a high CBX7 expression level predicts a favorable prognosis in meningioma patients. CBX7 restoration significantly induces cell cycle arrest and inhibits meningioma cell proliferation. iTRAQ-based proteomics analysis indicated that CBX7 restoration triggers the metabolic shift from glycolysis to oxidative phosphorylation. The mechanistic study demonstrated that CBX7 promotes the proteasome-dependent degradation of c-MYC proteins by transcriptionally inhibiting the expression of a c-MYC deubiquitinase, USP44, which attenuates c-MYC-mediated transactivation of LDHA transcripts and further inhibits glycolysis and subsequent cellular proliferation. More importantly, the functional role of CBX7 was further confirmed in both subcutaneous and orthotopic meningioma xenografts mouse models and human meningioma patients. Together, our results shed light on the critical role of CBX7 during meningioma malignancy progression and identified the CBX7/USP44/c-MYC/LDHA axis as a promising therapeutic target against meningioma progression.

17.
Cell Death Dis ; 13(6): 548, 2022 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-35697672

RESUMEN

Ferroptosis is a newly identified form of regulated cell death (RCD) characterized by the iron-dependent lipid reactive oxygen species (ROS) accumulation, but its mechanism in gliomas remains elusive. Acyl-coenzyme A (CoA) synthetase long-chain family member 4 (Acsl4), a pivotal enzyme in the regulation of lipid biosynthesis, benefits the initiation of ferroptosis, but its role in gliomas needs further clarification. Erastin, a classic inducer of ferroptosis, has recently been found to regulate lipid peroxidation by regulating Acsl4 other than glutathione peroxidase 4 (GPX4) in ferroptosis. In this study, we demonstrated that heat shock protein 90 (Hsp90) and dynamin-related protein 1 (Drp1) actively regulated and stabilized Acsl4 expression in erastin-induced ferroptosis in gliomas. Hsp90 overexpression and calcineurin (CN)-mediated Drp1 dephosphorylation at serine 637 (Ser637) promoted ferroptosis by altering mitochondrial morphology and increasing Acsl4-mediated lipid peroxidation. Importantly, promotion of the Hsp90-Acsl4 pathway augmented anticancer activity of erastin in vitro and in vivo. Our discovery reveals a novel and efficient approach to ferroptosis-mediated glioma therapy.


Asunto(s)
Ferroptosis , Glioma , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Dinaminas , Glioma/genética , Humanos , Lípidos , Serina
18.
Oncogene ; 41(18): 2597-2608, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35332268

RESUMEN

Glioblastoma (GBM) is the most aggressive primary brain tumor as one of the deadliest cancers. The TGF-ß signaling acts as an oncogenic factor in GBM, and plays vital roles in development of GBM. SMAD7 is a major inhibitor of TGF-ß signaling, while the deubiquitination of SMAD7 has been poorly studied in GBM. Here, we found USP2 as a new prominent candidate that could regulate SMAD7 stability. USP2 was lost in GBM, leading to the poor prognosis in patients. Moreover, aberrant DNA methylation mediated by DNMT3A induced the low expression of USP2 in GBM. USP2 depletion induced TGF-ß signaling and progression of GBM. In contrast, overexpressed USP2 suppressed TGF-ß signaling and GBM development. Specifically, USP2 interacted with SMAD7 and prevented SMAD7 ubiquitination. USP2 directly cleaved Lys27- and Lys48-linked poly-ubiquitin chains of SMAD7, and Lys27-linked poly-ubiquitin chains of SMAD7 K185 mediated the recruitment of SMAD7 to HERC3, which regulated Lys63-linked poly-ubiquitination of SMAD7. Moreover, we demonstrated that the DNMT3A inhibitor SGI-1027 induced USP2, suppressed TGF-ß signaling and GBM development. Thus, USP2 repressed development of GBM by inhibition TGF-ß signaling pathway via the deubiquitination of SMAD7.


Asunto(s)
Glioblastoma , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Poliubiquitina/metabolismo , Transducción de Señal , Proteína smad7/genética , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación
19.
Oxid Med Cell Longev ; 2021: 6338722, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34853630

RESUMEN

Traumatic brain injury (TBI) causes a high rate of mortality and disability, and its treatment is still limited. Loss of neurons in damaged area is hardly rescued by relative molecular therapies. Based on its disease characteristics, we transplanted human embryonic stem cell- (hESC-) derived cerebral organoids in the brain lesions of controlled cortical impact- (CCI-) modeled severe combined immunodeficient (SCID) mice. Grafted organoids survived and differentiated in CCI-induced lesion pools in mouse cortical tissue. Implanted cerebral organoids differentiated into various types of neuronal cells, extended long projections, and showed spontaneous action, as indicated by electromyographic activity in the grafts. Induced vascularization and reduced glial scar were also found after organoid implantation, suggesting grafting could improve local situation and promote neural repair. More importantly, the CCI mice's spatial learning and memory improved after organoid grafting. These findings suggest that cerebral organoid implanted in lesion sites differentiates into cortical neurons, forms long projections, and reverses deficits in spatial learning and memory, a potential therapeutic avenue for TBI.


Asunto(s)
Corteza Cerebral/patología , Organoides/trasplante , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones SCID , Transfección
20.
Neuro Oncol ; 23(12): 2014-2027, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33984142

RESUMEN

BACKGROUND: Ferroptosis, a programmed cell death characterized by lipid peroxidation, is implicated in various diseases including cancer. Although cell density-dependent E-cadherin and Merlin/Neurofibromin (NF2) loss can modulate ferroptosis, the role of ferroptosis and its potential link to NF2 status and E-cadherin expression in meningioma remain unknown. METHODS: Relationship between ferroptosis modulators expression and NF2 mutational status was examined in 35 meningiomas (10 NF2 loss and 25 NF2 wild type). The impact of NF2 and E-cadherin on ferroptosis were examined by lactate dehydrogenase (LDH) release, lipid peroxidation, and western blot assays in IOMM-Lee, CH157, and patient-derived meningioma cell models. Luciferase reporter and chromatin immunoprecipitation assays were used to assess the ability of MEF2C (myocyte enhancer factor 2C) to drive expression of NF2 and CDH1 (E-cadherin). Therapeutic efficacy of Erastin-induced ferroptosis was tested in xenograft mouse models. RESULTS: Meningioma cells with NF2 inactivation were susceptible to Erastin-induced ferroptosis. Meningioma cells grown at higher density increased expression of E-cadherin, which suppressed Erastin-induced ferroptosis. Maintaining NF2 and E-cadherin inhibited ferroptosis-related lipid peroxidation and meningioma cell death. MEF2C was found to drive the expression of both NF2 and E-cadherin. MEF2C silencing enhanced Erastin-induced ferroptotic meningioma cell death and lipid peroxidation levels in vitro, which was limited by forced expression of MEF2C targets, NF2 and E-cadherin. In vivo, anti-meningioma effect of Erastin was augmented by MEF2C knockdown and was counteracted by NF2 or E-cadherin. CONCLUSIONS: NF2 loss and low E-cadherin create susceptibility to ferroptosis in meningioma. MEF2C could be a new molecular target in ferroptosis-inducing therapies for meningioma.


Asunto(s)
Ferroptosis , Factores de Transcripción MEF2 , Neoplasias Meníngeas , Meningioma , Animales , Antígenos CD , Cadherinas/genética , Línea Celular Tumoral , Silenciador del Gen , Humanos , Factores de Transcripción MEF2/genética , Neoplasias Meníngeas/tratamiento farmacológico , Neoplasias Meníngeas/genética , Meningioma/tratamiento farmacológico , Meningioma/genética , Ratones , Neurofibromina 2 , Piperazinas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA