Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Front Oncol ; 11: 729103, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34568059

RESUMEN

Lower-grade glioma (LGG) is characterized by genetic and transcriptional heterogeneity, and a dismal prognosis. Iron metabolism is considered central for glioma tumorigenesis, tumor progression and tumor microenvironment, although key iron metabolism-related genes are unclear. Here we developed and validated an iron metabolism-related gene signature LGG prognosis. RNA-sequence and clinicopathological data from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) were downloaded. Prognostic iron metabolism-related genes were screened and used to construct a risk-score model via differential gene expression analysis, univariate Cox analysis, and the Least Absolute Shrinkage and Selection Operator (LASSO)-regression algorithm. All LGG patients were stratified into high- and low-risk groups, based on the risk score. The prognostic significance of the risk-score model in the TCGA and CGGA cohorts was evaluated with Kaplan-Meier (KM) survival and receiver operating characteristic (ROC) curve analysis. Risk- score distributions in subgroups were stratified by age, gender, the World Health Organization (WHO) grade, isocitrate dehydrogenase 1 (IDH1) mutation status, the O6-methylguanine-DNA methyl-transferase (MGMT) promoter-methylation status, and the 1p/19q co-deletion status. Furthermore, a nomogram model with a risk score was developed, and its predictive performance was validated with the TCGA and CGGA cohorts. Additionally, the gene set enrichment analysis (GSEA) identified signaling pathways and pathological processes enriched in the high-risk group. Finally, immune infiltration and immune checkpoint analysis were utilized to investigate the tumor microenvironment characteristics related to the risk score. We identified a prognostic 15-gene iron metabolism-related signature and constructed a risk-score model. High risk scores were associated with an age of > 40, wild-type IDH1, a WHO grade of III, an unmethylated MGMT promoter, and 1p/19q non-codeletion. ROC analysis indicated that the risk-score model accurately predicted 1-, 3-, and 5-year overall survival rates of LGG patients in the both TCGA and CGGA cohorts. KM analysis showed that the high-risk group had a much lower overall survival than the low-risk group (P < 0.0001). The nomogram model showed a strong ability to predict the overall survival of LGG patients in the TCGA and CGGA cohorts. GSEA analysis indicated that inflammatory responses, tumor-associated pathways, and pathological processes were enriched in high-risk group. Moreover, a high risk score correlated with the infiltration immune cells (dendritic cells, macrophages, CD4+ T cells, and B cells) and expression of immune checkpoint (PD1, PDL1, TIM3, and CD48). Our prognostic model was based on iron metabolism-related genes in LGG, can potentially aid in LGG prognosis, and provides potential targets against gliomas.

2.
Cell Commun Signal ; 19(1): 40, 2021 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-33761934

RESUMEN

BACKGROUND: Lysosome-associated membrane protein type 2A (LAMP-2A) is the key component of chaperone-mediated autophagy (CMA), a cargo-selective lysosomal degradation pathway. Aberrant LAMP-2A expression and CMA activation have been demonstrated in various human malignancies. The study focusing on the intrinsic role of LAMP-2A and CMA in glioblastoma (GBM), and downstream mechanism could provide valuable insight into the pathogenesis and novel therapeutic modality of GBM. METHODS: The levels of LAMP-2A, nuclear receptor co-repressor (N-CoR), unfolded protein response (UPR) and apoptosis were examined in clinical samples. LAMP-2A siRNA and shRNA were constructed to manipulate CMA activation. The role of CMA and downstream mechanism through degradation of N-CoR and arresting UPR mediated apoptosis were explored in GBM cells and nude mouse xenograft model. RESULTS: Elevated LAMP-2A and associated decreased N-CoR expression were observed in GBM as compared with peritumoral region and low-grade glioma. Inhibited UPR and apoptosis were observed in GBM with high LAMP-2A expression. In vitro study demonstrated co-localization and interaction between LAMP-2A and N-CoR. LAMP-2A silencing up-regulated N-CoR and aroused UPR pathway, leading to apoptosis, while N-CoR silencing led to an opposite result. In vivo study further confirmed that LAMP-2A inhibition arrested tumor growth by promoting apoptosis. CONCLUSIONS: Our results demonstrated the central role of CMA in mediating N-CoR degradation and protecting GBM cells against UPR and apoptosis, and provided evidence of LAMP-2A as potential biomarker. Further research focusing on CMA with other tumorigenic process is needed and selective modulators of LAMP-2A remain to be investigated to provide a novel therapeutic strategy for GBM. Video Abstract.


Asunto(s)
Apoptosis , Biomarcadores de Tumor/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Co-Represor 1 de Receptor Nuclear/metabolismo , Proteolisis , Animales , Apoptosis/genética , Caspasas/metabolismo , Línea Celular Tumoral , Autofagia Mediada por Chaperones/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Clasificación del Tumor , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Transcripción Genética , Respuesta de Proteína Desplegada/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Front Immunol ; 12: 617163, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33659003

RESUMEN

Preclinical and clinical research has demonstrated that inflammation is a critical factor regulating intracerebral hemorrhage (ICH)-induced brain injury. Growing evidence suggests that myeloid cells and lymphocytes have an effect on the pathophysiological processes associated with ICH, such as inflammation, immune responses, perihematomal edema formation, blood-brain barrier (BBB) integrity, and cell death. However, the underlying mechanisms remain largely unknown. We aimed to explore the role immune cells played at different stages of the ICH. To achieve this, novel bioinformatics algorithms were employed to analyze the gene expression profiles and three different analytical tools were utilized to predict the abundances of cell types. In this study, we found that natural killer (NK) cells infiltrated into the brain parenchyma after ICH. Infiltrating NK cells may mediate brain injury through degranulation and recruitment of other cells. Besides, in the acute phase of ICH, monocytes in peripheral blood carried out phagocytosis and secretion of cytokines. On the other hand, in the subacute stage, non-classical monocytes were activated and showed a stronger ability to carry out heme metabolism, wound healing, and antigen processing and presentation. In conclusion, our findings emphasize the significance of intracerebral infiltrating immunocytes in ICH and demonstrate that ICH is a systemic disease affected by peripheral blood. The hub genes identified might be promising therapeutic targets. We also provide a reference on how to use bioinformatics approaches to explore non-neoplastic immune-related diseases.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Leucocitos/metabolismo , Animales , Hemorragia Cerebral/etiología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Inmunohistoquímica , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucocitos/inmunología , Leucocitos/patología , Masculino , Ratones , Monocitos/inmunología , Monocitos/metabolismo
4.
J Neuroinflammation ; 18(1): 2, 2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33402181

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Microglial/macrophage activation and neuroinflammation are key cellular events following TBI, but the regulatory and functional mechanisms are still not well understood. Myeloid-epithelial-reproductive tyrosine kinase (Mer), a member of the Tyro-Axl-Mer (TAM) family of receptor tyrosine kinases, regulates multiple features of microglial/macrophage physiology. However, its function in regulating the innate immune response and microglial/macrophage M1/M2 polarization in TBI has not been addressed. The present study aimed to evaluate the role of Mer in regulating microglial/macrophage M1/M2 polarization and neuroinflammation following TBI. METHODS: The controlled cortical impact (CCI) mouse model was employed. Mer siRNA was intracerebroventricularly administered, and recombinant protein S (PS) was intravenously applied for intervention. The neurobehavioral assessments, RT-PCR, Western blot, magnetic-activated cell sorting, immunohistochemistry and confocal microscopy analysis, Nissl and Fluoro-Jade B staining, brain water content measurement, and contusion volume assessment were performed. RESULTS: Mer is upregulated and regulates microglial/macrophage M1/M2 polarization and neuroinflammation in the acute stage of TBI. Mechanistically, Mer activates the signal transducer and activator of transcription 1 (STAT1)/suppressor of cytokine signaling 1/3 (SOCS1/3) pathway. Inhibition of Mer markedly decreases microglial/macrophage M2-like polarization while increases M1-like polarization, which exacerbates the secondary brain damage and sensorimotor deficits after TBI. Recombinant PS exerts beneficial effects in TBI mice through Mer activation. CONCLUSIONS: Mer is an important regulator of microglial/macrophage M1/M2 polarization and neuroinflammation, and may be considered as a potential target for therapeutic intervention in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Polaridad Celular/fisiología , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Tirosina Quinasa c-Mer/biosíntesis , Animales , Lesiones Traumáticas del Encéfalo/prevención & control , Femenino , Mediadores de Inflamación/antagonistas & inhibidores , Activación de Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Front Cell Neurosci ; 14: 192, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32848609

RESUMEN

Background: Neuronal apoptosis involved in secondary injury following traumatic brain injury (TBI) significantly contributes to the poor outcomes of patients with TBI. The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can selectively induce apoptosis of tumor cells. Hypoxia factor (HIF) 1α is a controversial factor that mediates the neuronal apoptotic pathway. Herein, we hypothesize that HIF-1α may mediate the TRAIL-induced neuronal apoptosis after TBI. Methods: We used Western blots and immunofluorescence to study the expression and cell localization of TRAIL and death receptor 5 (DR5) after TBI in rats. Soluble DR5 (sDR5) administration was used to block the TRAIL-induced neuronal death and neural deficits. HIF-1α inhibitor 2ME and agonist DMOG were used to study the role of HIF-1α in TRAIL-induced neuronal death. Meanwhile, HIF-1α siRNA was used to investigate the role of HIF-1α in TRAIL-induced neuronal death in vitro. Results: The expressions of microglia-located TRAIL and neuron-located DR5 were significantly upregulated after TBI. sDR5 significantly attenuated TRAIL-induced neuronal apoptosis and neurological deficits. 2ME decreased neuronal apoptosis, lesion area, and brain edema and improved neurological function via increased expression of TRAIL decoy receptor 1 (DcR1), which inhibited TRAIL-induced apoptosis after TBI. The administration of DMOG produced the opposite effect than did 2ME. Similarly, HIF-1α siRNA attenuated TRAIL-induced neuronal death via increased DcR1 expression in vitro. Conclusion: Our findings suggested that the TRAIL/DR5 signaling pathway plays an important role after neuronal apoptosis after TBI. HIF-1α mediates TRAIL-induced neuronal apoptosis by regulating DcR1 expression following TBI.

6.
Front Immunol ; 10: 1360, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31258534

RESUMEN

Cell death is deeply involved in pathophysiology of brain injury after intracerebral hemorrhage (ICH). Necroptosis, one of the recently discovered forms of cell death, plays an important role in various diseases, including ICH. Previous studies have suggested that a considerable number of neurons undergoes necroptosis after ICH. However, necroptosis of microglia after ICH has not been reported to date. The present study demonstrated for the first time that necroptosis occurred in the microglia surrounding the hematoma after ICH in C57 mice, and melatonin, a hormone that is predominantly synthesized in and secreted from the pineal gland, exerted a neuroprotective effect by suppressing this process. When we further explored the potential underlying mechanism, we found that melatonin inhibits RIP3-mediated necroptosis by regulating the deubiquitinating enzyme A20 (also known as TNFAIP3) expression after ICH. In summary, we have demonstrated the role of microglial necroptosis in the pathogenesis of ICH. More importantly, A20 was identified as a novel target of melatonin, which opens perspectives for future research.


Asunto(s)
Lesiones Encefálicas/metabolismo , Hemorragia Cerebral/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Melatonina/metabolismo , Microglía/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis , Lesiones Encefálicas/patología , Hemorragia Cerebral/patología , Enzimas Desubicuitinizantes/genética , Modelos Animales de Enfermedad , Humanos , Inflamación , Ratones , Ratones Endogámicos C57BL , Necroptosis , Fármacos Neuroprotectores , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética
7.
World Neurosurg ; 101: 816.e5-816.e9, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28238872

RESUMEN

BACKGROUND: Intracavernous internal carotid artery (ICICA) aneurysm secondary to pituitary infection is exceedingly rare. CASE DESCRIPTION: We report an unusual case of a 63-year-old man who presented with acute left blepharoptosis and imaging findings of a pituitary infection. Interestingly, sudden onset of right blepharoptosis occurred after anti-infective therapy for 10 days. Digital subtraction angiography revealed a right ICICA aneurysm. After 6 months of follow-up visits, enlargement of the ICICA aneurysm was observed, and the endovascular technique of a low-profile visualized intraluminal support stent combined with 5 detachable coils was successfully performed to treat the ICICA aneurysm, with preservation of the internal carotid artery. CONCLUSION: This rare case highlights a life-threatening complication of a pituitary infection. Moreover, enlargement of the infected ICICA aneurysm could not be relieved by conservative anti-infective therapy. Endovascular treatment may be an alternative therapy for an infected ICICA aneurysm.


Asunto(s)
Aneurisma Infectado/cirugía , Enfermedades de las Arterias Carótidas/cirugía , Procedimientos Endovasculares , Aneurisma Intracraneal/cirugía , Leucocitosis/cirugía , Enfermedades de la Hipófisis/cirugía , Aneurisma Infectado/diagnóstico por imagen , Aneurisma Infectado/etiología , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/etiología , Arteria Carótida Interna/diagnóstico por imagen , Arteria Carótida Interna/cirugía , Procedimientos Endovasculares/métodos , Humanos , Aneurisma Intracraneal/diagnóstico por imagen , Aneurisma Intracraneal/etiología , Leucocitosis/complicaciones , Leucocitosis/diagnóstico por imagen , Masculino , Persona de Mediana Edad , Enfermedades de la Hipófisis/complicaciones , Enfermedades de la Hipófisis/diagnóstico por imagen , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA