Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cell Mol Life Sci ; 81(1): 247, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38829550

RESUMO

BACKGROUND: The high degree of intratumoral genomic heterogeneity is a major obstacle for glioblastoma (GBM) tumors, one of the most lethal human malignancies, and is thought to influence conventional therapeutic outcomes negatively. The proneural-to-mesenchymal transition (PMT) of glioma stem cells (GSCs) confers resistance to radiation therapy in glioblastoma patients. POLD4 is associated with cancer progression, while the mechanisms underlying PMT and tumor radiation resistance have remained elusive. METHOD: Expression and prognosis of the POLD family were analyzed in TCGA, the Chinese Glioma Genome Atlas (CGGA) and GEO datasets. Tumorsphere formation and in vitro limiting dilution assay were performed to investigate the effect of UCHL3-POLD4 on GSC self-renewal. Apoptosis, TUNEL, cell cycle phase distribution, modification of the Single Cell Gel Electrophoresis (Comet), γ-H2AX immunofluorescence, and colony formation assays were conducted to evaluate the influence of UCHL3-POLD4 on GSC in ionizing radiation. Coimmunoprecipitation and GST pull-down assays were performed to identify POLD4 protein interactors. In vivo, intracranial xenograft mouse models were used to investigate the molecular effect of UCHL3, POLD4 or TCID on GCS. RESULT: We determined that POLD4 was considerably upregulated in MES-GSCs and was associated with a meagre prognosis. Ubiquitin carboxyl terminal hydrolase L3 (UCHL3), a DUB enzyme in the UCH protease family, is a bona fide deubiquitinase of POLD4 in GSCs. UCHL3 interacted with, depolyubiquitinated, and stabilized POLD4. Both in vitro and in vivo assays indicated that targeted depletion of the UCHL3-POLD4 axis reduced GSC self-renewal and tumorigenic capacity and resistance to IR treatment by impairing homologous recombination (HR) and nonhomologous end joining (NHEJ). Additionally, we proved that the UCHL3 inhibitor TCID induced POLD4 degradation and can significantly enhance the therapeutic effect of IR in a gsc-derived in situ xenograft model. CONCLUSION: These findings reveal a new signaling axis for GSC PMT regulation and highlight UCHL3-POLD4 as a potential therapeutic target in GBM. TCID, targeted for reducing the deubiquitinase activity of UCHL3, exhibited significant synergy against MES GSCs in combination with radiation.


Assuntos
Células-Tronco Neoplásicas , Tolerância a Radiação , Ubiquitina Tiolesterase , Humanos , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/genética , Tolerância a Radiação/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Animais , Camundongos , Linhagem Celular Tumoral , Glioma/patologia , Glioma/genética , Glioma/radioterapia , Glioma/metabolismo , Apoptose/genética , Apoptose/efeitos da radiação , Ubiquitinação , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Camundongos Nus , Fenótipo , Regulação Neoplásica da Expressão Gênica , Prognóstico
2.
Cancer Cell Int ; 21(1): 347, 2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34225723

RESUMO

BACKGROUND: Glioblastoma remains one of the most lethal brain cancers. T-cell immunoglobulin and mucin domain 1 (Tim-1) is associated with various immune diseases. The molecular mechanism of Tim-1 in regulating glioblastoma cell proliferation, invasion, and migration is still unknown. Moreover, it has shown that miR-133a plays an important role in glioblastoma. However, little is known about the interaction between Tim-1 and miR-133a in glioblastoma. METHODS: Tim-1 expression in glioblastoma and normal brain tissues was detected by qPCR, Western Blot and IHC. After Tim-1 knockdown in U251 and U87 cells, genes showing significantly differential expression, along with the significant differential miRNAs were analyzed using RNA-seq analysis. The binding sites were verified using dual-luciferase reporter gene assay. U251 and U87 cells were allocated into the small harpin-negative control (sh-NC), sh-Tim-1, sh-Tim-1 + inhibitor NC, and sh-Tim-1 + miR-133a inhibitor group. Cell proliferation, migration, and invasion were determined by CCK-8, flow cytometry, wound-healing and Transwell assays, respectively. Next, U251 and U87 cells were allocated into the mimic NC, miR-133a mimic, miR-133a mimic + pcDNA3.1, and miR-133a mimic + pcDNA3.1-TGFBR1 groups, followed by the detection of cell proliferation, migration, and invasion. Western blot was used to identify the expression of vital kinases in the Wnt/ß-catenin pathway. RESULTS: Tim-1 was highly expressed in glioblastoma tissues compared with that in normal brain tissues. RNA-seq analysis showed that Tim-1 knockdown could lead to the downregulation of TGFBR1 and the upregulation of miR-133a. The binding sites between TGFBR1 and miR-133a were confirmed. Tim-1 knockdown impaired the invasion, migration, proliferation of U251 and U87 cells, which could be reversed by miR-133a downregulation. miR-133a upregulation inhibited the proliferation, invasion, and migration of U251 and U87 cells, which could be reversed by TGFBR1 upregulation. Tim-1 knockdown and miR-133a upregulation could inhibit the activation of the Wnt/ß-catenin pathway, while the elevation of TGFBR1 showed opposite effects. CONCLUSION: Tim-1 knockdown inhibited glioblastoma cell proliferation, invasion, and migration through the miR-133a/TGFBR1 axis and restrained the activation of the Wnt/ß-catenin pathway.

3.
Neoplasma ; 68(4): 732-741, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33847130

RESUMO

Protein- or peptide-based therapeutics have emerged as an innovative strategy for the treatment of cancer. Our previous research demonstrated that tripartite motif 9 short isoform (TRIM9s) is a tumor suppressor in glioma. In this report, we investigated whether a new peptide derived from TRIM9s, named T9sP, inhibits glioma progression and determined the possible molecular mechanism. The CCK-8 proliferation assay was performed in LN229 and U251 glioma cells. The scratch-wound assay was used to determine the migration of the cells. Apoptosis was assessed by flow cytometry using Annexin V-FITC/PI double staining method. The relative protein expression levels were detected by immunoblot analysis. The cell-penetrating peptide TAT was fused with T9sP to form TAT-T9sP. TAT-T9sP efficiently penetrated through the cell membrane of both LN229 and U251 cells. TAT-T9sP inhibited proliferation and migration and promoted apoptosis of glioma cells. TAT-T9sP activated p38 signaling by upregulating MKK6, and a p38 inhibitor, SB203580, reversed the inhibitory effects of TAT-T9sP on glioma cells. These results indicated the potential of TAT-T9sP for the development of a new anti-glioma medicine.


Assuntos
Neoplasias Encefálicas , Glioma , Apoptose , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Glioma/tratamento farmacológico , Humanos , Peptídeos/farmacologia , Transdução de Sinais
4.
J Craniofac Surg ; 32(1): e19-e20, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32858607

RESUMO

OBJECTIVES: The aim of this study was to study and explore the genetic mechanism of familial meningiomas through 3 cases of familial tuberculum sellae meningioma. METHODS: A retrospective analysis of clinical data of 3 cases of familial tuberculum sellae meningioma patients, and the pathological results of types and immunohistochemical results of the 3 patients were compare. REULTS: Three cases of postoperative pathology were meningiomas (mixed type), immunohistochemical examination showed that Vimentin, epithelial membrain antigen , and Ki67 were positive. CONCLUSIONS: The occurrence of meningiomas is associated with ≥1 chromosomal deletions, and the absence of certain tumor suppressor genes may be the genetic basis for the familial growth of meningiomas.


Assuntos
Neoplasias Meníngeas , Meningioma , Neoplasias da Base do Crânio , Idoso , Feminino , Humanos , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/cirurgia , Meningioma/genética , Meningioma/cirurgia , Procedimentos Neurocirúrgicos , Estudos Retrospectivos , Sela Túrcica , Neoplasias da Base do Crânio/cirurgia , Resultado do Tratamento
5.
Cancer Cell Int ; 19: 280, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31708690

RESUMO

BACKGROUND: Glioma is the most common central nervous system (CNS) tumour. p62, an important autophagy adaptor, plays a crucial role in cancer. However, the role of p62 in the progression of glioma is poorly characterized. METHODS: We examined the expression of p62 in glioma tissues and cell lines. Then we investigated the function of p62 in vitro, and clarified the mechanism underlying the regulation of p62 expression. RESULTS: We revealed that p62 was upregulated at both the mRNA and protein levels in human glioma tissues irrelevant to isocitrate dehydrogenase (IDH) status. Then, we found that overexpression of p62 promoted glioma progression by promoting proliferation, migration, glycolysis, temozolomide (TMZ) resistance and nuclear factor κB (NF-κB) signalling pathway, and repressing autophagic flux and reactive oxygen species (ROS) in vitro. In accordance with p62 overexpression, knockdown of p62 exerted anti-tumour effects in glioma cells. Subsequently, we demonstrated that miR-124-3p directly targeted the 3'-UTR of p62 mRNA, leading to the downregulation of p62. Finally, we found that p62 function could be partially reversed by miR-124-3p overexpression. CONCLUSIONS: Our results demonstrate that p62 can be targeted by miR-124-3p and acts as an oncogene in glioma, suggesting the potential value of p62 as a novel therapeutic target for glioma.

6.
J Neurooncol ; 141(2): 277-288, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30460630

RESUMO

OBJECTIVE: Malignant glioma is a lethal brain tumor with a low survival rate and poor prognosis. New strategies are urgently needed to augment the chemotherapeutic effects of temozolomide (TMZ), the standard drug in glioma treatment. Carnosic acid (CA) has been reported to have anticancer, antioxidant and anti-infectious properties. In this study, we aimed to investigate the anticancer effects and the underlying mechanisms of CA in combination with TMZ in glioma cancer cells. METHODS: The glioma cancer cells were treated with TMZ, CA, or TMZ + CA. We evaluated cell survival by CCK-8 assay, cell anchorage-independent survival by colony formation assay, cell migration by wound-healing assay, cell cycle and cell apoptosis by flow cytometry, and protein expression by western blot. RESULTS: CA enhanced the cytotoxic effect of TMZ in glioma cancer cells. CA enhanced TMZ-induced inhibition of colony formation and cell migration and enhanced TMZ-induced cell cycle arrest and cellular apoptosis. Immunofluorescence suggested that CA in combination with TMZ triggered autophagy. Furthermore, CA promoted TMZ-induced cell cycle arrest and cellular apoptosis by Cyclin B1 inhibition and activation of PARP and Caspase-3, while CA promoted TMZ-induced cellular autophagy by p-AKT inhibition, p62 downregulation and LC3-I to LC3-II transition. CONCLUSION: These data suggest that the combination therapy of CA and TMZ strengthens the anticancer effect of TMZ by enhancing apoptosis and autophagy.


Assuntos
Abietanos/administração & dosagem , Antineoplásicos Alquilantes/administração & dosagem , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Temozolomida/administração & dosagem , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Humanos
7.
Cell Physiol Biochem ; 48(2): 450-460, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30016785

RESUMO

BACKGROUND/AIMS: Glioma causes significant human mortalities annually. Molecularly-targeted therapy is a focus of glioma research. METHODS: Grb2-associated binding 1 (Gab1) expression and microRNA-29a-3p ("miR-29a-3p") expression in human glioma cells and tissues were tested by Western blotting assay and qRT-PCR assay. shRNA/siRNA strategy was applied to silence Gab1 in human glioma cells. miR-29a or anti-sense miR-29a construct was transfected to human glioma cells. Cell proliferation was tested by BrdU ELISA assay and cell counting assay. RESULTS: We show that expression of Gab1 was significantly elevated in human glioma tissues and cells, which correlated with downregulation of its putative microRNA: miR-29a-3p. In A172 glioma cells and primary human glioma cells, Gab1 shRNA/siRNA inhibited Akt-Erk activation and cell proliferation. Forced-expression of miR-29a-3p downregulated Gab1, inhibiting glioma cell proliferation, whereas miR-29a-3p was in-effective on cell proliferation in Gab1-silenced A172 cells. Furthermore, introduction of a 3'-untranslated region (3'-UTR) mutant Gab1 (UTR-G160A) blocked miR-29a-3p-induced inhibition on Akt signaling and A172 cell proliferation. CONCLUSIONS: miR-29a-3p downregulation leads to Gab1 upregulation to promote glioma cell proliferation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Glioma/patologia , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Antagomirs/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioma/metabolismo , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
8.
Cell Physiol Biochem ; 44(6): 2296-2309, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29262398

RESUMO

BACKGROUND/AIMS: Hypoxic/ischemic injury to the liver is a frequently encountered clinical problem with limited therapeutic options. Since microRNAs (miRNAs) are involved in hypoxic/ ischemic events, and δ-opioid receptor (DOR) is protective against hypoxic/ischemic injury, we asked if pharmacological activation of DOR can alter hypoxic events by regulating miRNA expression in the liver. As the first step, the present work aimed at testing the effect of DOR activation on hepatic miRNA expression in hypoxia. METHODS: Male Sprague Dawley rats were exposed to hypoxia (9.5-10% O2) for 1, 5, or 10 days with or without DOR activation. The target miRNAs were selected according to TaqMan low-density array (TLDA) data and analyzed by quantitative real-time PCR. RESULTS: We found that: 1) 1-day hypoxia caused the upregulation of 9 miRNAs (miR-7a-5p, miR-10a-5p, miR-25-3p, miR-26b-5p, miR-122-5p, miR-128a-3p, miR-135b-5p, miR-145-5p, and miR-181a-5p) and the downregulation of 2 miRNAs (miR-34a-5p and miR-182); 2) 5 and 10-days hypoxia altered the expression of 4 miRNAs (miR-34c-5p, miR-184, miR-107-3p and miR192-5p); 3) DOR activation shifted the expression of 8 miRNAs (miR-122-5p, miR-146a-5p, miR-30e-5p, miR-128a-3p, miR-182, miR-192-5p miR-107-3p and miR-184) in normoxic condition; and 4) DOR activation modified hypoxia-induced changes in 6 miRNAs (miR-142-5p, miR-145-5p, miR-146a-5p, miR-204-5p, miR-34a-5p and miR-192-5p). CONCLUSION: Hypoxia significantly modifies the miRNA profile in the liver, while DOR activation can modify the hypoxic modification. Therefore, it is potentially possible to alter hypoxic/ischemic pathophysiology in the liver through DOR pharmacotherapy.


Assuntos
Regulação da Expressão Gênica , Hipóxia/genética , Fígado/metabolismo , MicroRNAs/genética , Receptores Opioides delta/genética , Animais , Perfilação da Expressão Gênica , Hipóxia/patologia , Fígado/patologia , Masculino , Ratos Sprague-Dawley
10.
J Stroke Cerebrovasc Dis ; 26(10): 2376-2382, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28666803

RESUMO

BACKGROUND: Recently, favorable outcomes from several randomized controlled trials of rapid endovascular treatment for acute ischemic stroke has emerged. OBJECTIVE: The aim of this retrospective study is to present our clinical experience in severe acute vertebrobasilar occlusion (AVBO) using intra-arterial treatment (IAT). METHODS: Twenty patients with ischemic stroke in the vertebrobasilar circulation treated by IAT between March 2011 and December 2014 were included. We retrospectively assessed National Institutes of Health Stroke Scale (NIHSS) score on admission and at discharge, Thrombolysis in Cerebral Infarction (TICI) scale, and clinical outcome using modified Rankin scale (mRs) at 90 days, and causes of stroke were prospectively assessed. RESULTS: The mean NIHSS score on admission was 26.4 ± 7.9 (range 9-33) points. The mean time from symptom onset to revascularization was 349.5 ± 124.0 (range 201-579) minutes. Successful recanalization (TICI ≥2b) was achieved in 19 (95.0%) patients. The mean NIHSS score at discharge was 5.7 ± 9.0 (range 0-30) points. A favorable clinical outcome (mRS ≤2) was observed in 12 (60.0%) patients at 90 days and mortality was 25.0% (n = 5). CONCLUSION: IAT for AVBO provides high rate of recanalization, favorable clinical outcome, and improved survival.


Assuntos
Isquemia Encefálica/terapia , Revascularização Cerebral , Trombólise Mecânica , Stents , Acidente Vascular Cerebral/terapia , Adulto , Idoso , Artéria Basilar/diagnóstico por imagem , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/mortalidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Índice de Gravidade de Doença , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/mortalidade , Resultado do Tratamento , Artéria Vertebral/diagnóstico por imagem
11.
Cancer Sci ; 107(7): 899-907, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27088547

RESUMO

The PIM1 protein is an important regulator of cell proliferation, the cell cycle, apoptosis, and metabolism in various human cancers. MicroRNAs (miRNAs) are powerful post-transcriptional gene regulators that function through translational repression or transcript destabilization. Therefore, we aimed to identify whether a close relationship exists between PIM1 and miRNAs. PIM1 protein levels and mRNA levels were significantly upregulated in astrocytoma tissues, indicating the oncogenic role of PIM1 in astrocytoma. Further bioinformatics analysis indicated that miR-124-3p targeted the 3'-UTR of PIM1. We also observed an inverse correlation between the miR-124-3p levels and PIM1 protein or mRNA levels in astrocytoma samples. Next, we experimentally confirmed that miR-124-3p directly recognizes the 3'-UTR of the PIM1 transcript and regulates PIM1 expression at both the protein and mRNA levels. Furthermore, we examined the biological consequences of miR-124-3p targeting PIM1 in vitro. We showed that the repression of PIM1 in astrocytoma cancer cells by miR-124-3p suppressed proliferation, invasion, and aerobic glycolysis and promoted apoptosis. We observed that the restoration or inhibition of PIM1 activity resulted in effects that were similar to those induced by miR-124-3p inhibitors or mimics in cancer cells. Finally, overexpression of PIM1 rescued the inhibitory effects of miR-124-3p. In summary, these findings aid in understanding the tumor-suppressive role of miR-124-3p in astrocytoma pathogenesis through the inhibition of PIM1 translation.


Assuntos
Apoptose/genética , Astrocitoma/genética , Astrocitoma/patologia , Metabolismo Energético/genética , MicroRNAs/genética , Invasividade Neoplásica/genética , Proteínas Proto-Oncogênicas c-pim-1/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Regiões 3' não Traduzidas/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-pim-1/biossíntese , Regulação para Cima
12.
Cell Physiol Biochem ; 39(3): 1118-28, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27576463

RESUMO

BACKGROUND: Hypoxic/ischemic injury to the heart is a frequently encountered clinical problem with limited therapeutic options. Since microRNAs (miRNAs) are involved in hypoxic/ischemic events, and δ-opioid receptor (DOR) activation is known to protect against hypoxic/ischemic injury, we speculated on the involvement of DOR activation in altering miRNA expression in the heart under hypoxic conditions. The present study aimed to test our hypothesis. METHODS: Male Sprague Dawley rats were exposed to hypoxia (9.5-10% O2) for 1, 5, or 10 days with or without DOR activation. The target miRNAs were selected from TaqMan low-density array (TLDA) data and were further analyzed by quantitative real-time PCR. RESULTS: We found that: 1) hypoxia alters the miRNA expression profiles depending on the hypoxic duration; 2) DOR activation shifts miRNA expression profiles in normoxic conditions and upregulates miR-128a-3p, miR-134-5p, miR-135a, miR-193a-3p, miR-196a, miR-324-3p, and miR-338; and 3) DOR activation modifies hypoxia-induced changes in miRNA expression and increases the levels of miR-128a-3p, miR-134-5p, miR-135a, miR-193a-3p, miR-196a, miR-324-3p, miR-141, miR-200b, and miR-324-3p. For example, miR-196c-5p decreased by 50% while miR-135a-5p increased 2.9 fold after 10 days under hypoxic conditions. Moreover, DOR activation further strengthened the hypoxia-induced increase of the levels of miR-7a-5p. When DOR was activated using UFP-512, the level of miR-107-3p significantly increased 1 day after the administration of UFP-512, but gradually decreased back to normal under normoxia. CONCLUSION: Hypoxia significantly modifies the miRNA profile in the heart, which can be mimicked or modified by DOR activation. Defining the targeted pathways that regulate the diverse cellular and molecular functions of miRNAs may provide new insights into potential therapies for hypoxic/ischemic injury of the heart.


Assuntos
Hipóxia/genética , MicroRNAs/genética , Isquemia Miocárdica/genética , Miocárdio/metabolismo , Receptores Opioides delta/genética , Animais , Benzimidazóis/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hipóxia/metabolismo , Hipóxia/patologia , Masculino , MicroRNAs/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Transdução de Sinais
13.
Tumour Biol ; 37(3): 3331-40, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26440052

RESUMO

Astrocytoma is one of the most common primary central nervous system tumors and has both high mortality and a poor 5-year survival rate. MicroRNAs (miRNAs) play important roles in carcinogenesis by acting on multiple signaling pathways. Although we have demonstrated that miR-137 is downregulated in astrocytoma tissues, the role of miR-137 in astrocytoma still remains unknown. In the present study, we aimed to investigate the function of miR-137 and its possible target genes in astrocytoma. miR-137 was significantly downregulated in astrocytoma tissues, and its expression level was inversely correlated with the clinical stage. Restoring miR-137 was able to dramatically inhibit cell proliferation, migration, and invasion and enhance apoptosis in vitro, whereas silencing its expression inhibited these processes. By overexpressing or inhibiting miR-137 in cancer cells, we experimentally confirmed that miR-137 directly recognized the 3'-UTR (3'-untranslated region) of the RASGRF1 (Ras protein-specific guanine nucleotide-releasing factor 1) transcript and regulated RASGRF1 expression. Furthermore, an inverse correlation was observed between miR-137 levels and RASGRF1 protein levels, but not mRNA levels, in astrocytoma samples. The silencing of RASGRF1 resulted in similar effects to miR-137 restoration in cancer cells. Finally, overexpression of RASGRF1 rescued the inhibitory effects of miR-137. Taken together, our results indicate that miR-137 acts as a tumor suppressor in astrocytoma by targeting RASGRF1. These findings suggest that miR-137 may serve as a novel therapeutic target in astrocytoma treatment.


Assuntos
Regiões 3' não Traduzidas/genética , Astrocitoma/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , ras-GRF1/genética , Apoptose/genética , Astrocitoma/metabolismo , Astrocitoma/patologia , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo , Genes Supressores de Tumor , Humanos , Invasividade Neoplásica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , ras-GRF1/metabolismo
14.
Int J Mol Sci ; 17(8)2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27517901

RESUMO

The pathological changes of Parkinson's disease (PD) are, at least partially, associated with the dysregulation of PTEN-induced putative kinase 1 (PINK1) and caspase 3. Since hypoxic and neurotoxic insults are underlying causes of PD, and since δ-opioid receptor (DOR) is neuroprotective against hypoxic/ischemic insults, we sought to determine whether DOR activation could protect the cells from damage induced by hypoxia and/or MPP⁺ by regulating PINK1 and caspase 3 expressions. We exposed PC12 cells to either severe hypoxia (0.5%-1% O2) for 24-48 h or to MPP⁺ at different concentrations (0.5, 1, 2 mM) and then detected the levels of PINK1 and cleaved caspase 3. Both hypoxia and MPP⁺ reduced cell viability, progressively suppressed the expression of PINK1 and increased the cleaved caspase 3. DOR activation using UFP-512, effectively protected the cells from hypoxia and/or MPP⁺ induced injury, reversed the reduction in PINK1 protein and significantly attenuated the increase in the cleaved caspase 3. On the other hand, the application of DOR antagonist, naltrindole, greatly decreased cell viability and increased cleaved caspase 3. These findings suggest that DOR is cytoprotective against both hypoxia and MPP⁺ through the regulation of PINK1 and caspase 3 pathways.


Assuntos
Caspase 3/metabolismo , Receptores Opioides delta/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células PC12 , Doença de Parkinson/metabolismo , Proteínas Quinases/metabolismo , Ratos
15.
Neurol Sci ; 36(2): 309-13, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25190548

RESUMO

microRNAs (miRNAs) are a class of small non-coding RNAs, approximately 21-25 nucleotides in length. Recently, some researchers have demonstrated that plasma miRNAs are sensitive and specific biomarkers of various cancers. Using quantitative real-time PCR, the expression levels of miR-454-3p were compared between pre-operative plasmas from 70 glioma patients and 70 healthy controls, and between these pre-operative and post-operative plasmas. Kaplan-Meier analysis was used to evaluate the association of miR-454-3p with prognosis of glioma patients. The expression levels of miR-454-3p in plasma in glioma patients were significantly higher than that from healthy controls. The expression levels of miR-454-3p were higher in high grade gliomas than in low grade gliomas. The AUC of the expression of miR-454-3p in plasma for glioma diagnosis was 0.9063. The expression levels of miR-454-3p in the post-operative plasmas were significantly downregulated when compared to the pre-operative plasmas. Moreover, the prognosis of glioma with high miR-454-3p expression was significantly worse compared with that of glioma with low miR-454-3p expression. Our results suggested that plasma miR-454-3p could be a novel potential diagnostic biomarker for glioma.


Assuntos
Neoplasias Encefálicas/sangue , Glioma/sangue , MicroRNAs/sangue , Área Sob a Curva , Biomarcadores Tumorais/sangue , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/diagnóstico , Glioma/patologia , Glioma/cirurgia , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
J Surg Res ; 191(1): 156-60, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24888789

RESUMO

BACKGROUND: Epidemiological studies have been conducted to investigate the association of telomerase reverse transcriptase (TERT) rs2736100 polymorphism with glioma risk. The aim of the present study was to evaluate the association of TERT rs2736100 polymorphism with glioma risk using a meta-analysis approach. MATERIALS AND METHODS: All eligible studies were identified through a search of PubMed, EMBASE, China National Knowledge Infrastructure, Database of Chinese Scientific and Technical Periodicals, and China Biology Medical literature database before January 2014. The association between the TERT rs2736100 polymorphism and glioma risk was estimated by odds ratio (OR) and 95% confidence interval (CI). RESULTS: A total of nine case-control studies including 9411 cases and 13,708 controls were eventually collected. Overall, we found that TERT rs2736100 polymorphism was significantly associated with the risk of glioma (OR = 1.29, 95% CI 1.24-1.34, P < 0.001). In the subgroup analysis based on ethnicity, the significant association was found in Caucasians (OR = 1.29, 95% CI 1.24-1.34, P < 0.001). In subgroup analyses by histology, the associations were significant in glioblastoma (OR = 1.45, 95% CI 1.32-1.60, P < 0.001), astrocytoma (OR = 1.41, 95% CI 1.26-1.58, P < 0.001), and oligodendroglioma (OR = 1.20, 95% CI 1.05-1.37, P = 0.008). CONCLUSIONS: Taken together, these data suggested that TERT rs2736100 polymorphism may contribute to glioma susceptibility.


Assuntos
Neoplasias Encefálicas/etnologia , Neoplasias Encefálicas/genética , Glioma/etnologia , Glioma/genética , Polimorfismo Genético , Telomerase/genética , Povo Asiático/genética , Povo Asiático/estatística & dados numéricos , Astrocitoma/etnologia , Astrocitoma/genética , Predisposição Genética para Doença/etnologia , Predisposição Genética para Doença/genética , Glioblastoma/etnologia , Glioblastoma/genética , Humanos , Oligodendroglioma/etnologia , Oligodendroglioma/genética , Fatores de Risco , População Branca/genética , População Branca/estatística & dados numéricos
17.
PLoS One ; 19(7): e0306667, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38950023

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0109124.].

18.
Cancer Lett ; 593: 216875, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38643837

RESUMO

Mesenchymal glioma stem cells (MES GSCs) are a subpopulation of cells in glioblastoma (GBM) that contribute to a worse prognosis owing to their highly aggressive nature and resistance to radiation therapy. Here, OCT4 is characterized as a critical factor in sustaining the stemness phenotype of MES GSC. We find that OCT4 is expressed intensively in MES GSC and is intimately associated with poor prognosis, moreover, OCT4 depletion leads to diminished invasive capacity and impairment of the stem phenotype in MES GSC. Subsequently, we demonstrated that USP5 is a deubiquitinating enzyme which directly interacts with OCT4 and preserves OCT4 stability through its deubiquitination. USP5 was additionally proven to be aberrantly over-expressed in MES GSCs, and its depletion resulted in a noticeable diminution of OCT4 and consequently a reduced self-renewal and tumorigenic capacity of MES GSCs, which can be substantially restored by ectopic expression of OCT4. In addition, we detected the dominant molecule that regulates USP5 transcription, E2F1, with dual luciferase reporter gene analysis. In combination, targeting the E2F1-USP5-OCT4 axis is a potentially emerging strategy for the therapy of GBM.


Assuntos
Neoplasias Encefálicas , Fator de Transcrição E2F1 , Células-Tronco Neoplásicas , Fator 3 de Transcrição de Octâmero , Proteases Específicas de Ubiquitina , Humanos , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/metabolismo , Animais , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F1/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo , Glioma/patologia , Glioma/genética , Glioma/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Camundongos , Estabilidade Proteica , Glioblastoma/patologia , Glioblastoma/genética , Glioblastoma/metabolismo , Ubiquitinação
19.
Int J Cancer ; 132(1): 128-36, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22674195

RESUMO

The aberrant expression of microRNAs (miRNAs) is associated with a variety of diseases, including cancer. In our study, we examined the miRNA expression profile of meningiomas, which is a common type of benign intracranial tumor derived from the protective meninges membranes that surround the brain and spinal cord. To define a typical human meningioma miRNA profile, the expression of 200 miRNAs in a training sample set were screened using quantitative reverse transcription polymerase chain reaction analysis, and then significantly altered miRNAs were validated in a secondary independent sample set. Kaplan-Meier and univariate/multivariate Cox proportional hazard regression analyses were performed to assess whether miRNA expression could predict the recurrence of meningioma after tumor resection. After a two-phase selection and validation process, 14 miRNAs were found to exhibit significantly different expression profiles in meningioma samples compared to normal adjacent tissue (NAT) samples. Unsupervised clustering analysis indicated that the 14-miRNA profile differed between tumor and NAT samples. Downregulation of miR-29c-3p and miR-219-5p were found to be associated with advanced clinical stages of meningioma. Kaplan-Meier analysis showed that high expression of miR-190a and low expression of miR-29c-3p and miR-219-5p correlated significantly with higher recurrence rates in meningioma patients. Cox proportional hazard regression analysis revealed that miR-190a expression level is an important prognostic predictor that is independent of other clinicopathological factors. Our results suggest that the use of miRNA profiling has significant potential as an effective diagnostic and prognostic marker in defining the expression signature of meningiomas and in predicting postsurgical outcomes.


Assuntos
Neoplasias Meníngeas/genética , Meningioma/genética , Meningioma/patologia , MicroRNAs/biossíntese , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Adolescente , Adulto , Idoso , Criança , Análise por Conglomerados , Regulação para Baixo , Feminino , Seguimentos , Perfilação da Expressão Gênica/métodos , Humanos , Estimativa de Kaplan-Meier , Masculino , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patologia , Meningioma/metabolismo , MicroRNAs/genética , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Prognóstico , Modelos de Riscos Proporcionais , Adulto Jovem
20.
Oper Neurosurg (Hagerstown) ; 22(5): 290-297, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35315837

RESUMO

BACKGROUND: The management of petroclival region meningioma remains the ultimate achievement in neurosurgery, because of the formidable technical challenges involved. OBJECTIVE: To describe the technique and feasibility of the purely endoscopic far-lateral supracerebellar infratentorial approach (EF-SCITA) for the treatment of petroclival region meningiomas. METHODS: We reviewed the clinical data of 10 consecutive cases of petroclival region meningiomas treated with the EF-SCITA from August 2018 to August 2020. The clinical outcomes were analyzed. The patient was placed in the lateral position, and then, a "C" shaped incision and craniotomy with exposed sigmoid and transverse sinuses were performed. With the endoscopic holder, endoscopic procedures were performed using standard 2-hand microsurgical techniques. Whether the tentorium or Meckel cave was handled depended on the tumor extension. RESULTS: The mean diameter was 45 × 25 mm. Dizziness and headache were the main symptoms. All 10 patients achieved gross total or subtotal resection (Petroclival Meningioma Grade I-III) with good neurological outcomes. The EF-SCITA provides satisfactory, direct exposure to the petroclival region. Cranial nerve deficits are the main postoperative complications. Two patients had a trochlear nerve injury, 3 patients had transient facial paralysis, and 2 patients had oculomotor paralysis (1 total and 1 incomplete), but both of them recovered during the follow-up period. One patient experienced an ipsilateral superior cerebellar artery infarction, and another patient had transient hemiparesis. CONCLUSION: The EF-SCITA is effective for most petroclival region meningiomas, except for the cavernous sinus type. This approach simplifies craniotomy procedures, omits burdensome petrosectomy, and avoids crossing posterior neurovascular structures.


Assuntos
Neoplasias Meníngeas , Meningioma , Neoplasias da Base do Crânio , Craniotomia/métodos , Humanos , Neoplasias Meníngeas/diagnóstico por imagem , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/cirurgia , Meningioma/diagnóstico por imagem , Meningioma/patologia , Meningioma/cirurgia , Procedimentos Neurocirúrgicos/métodos , Neoplasias da Base do Crânio/cirurgia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa