Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Lett ; 592: 216927, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38697460

RESUMO

Glioblastoma (GBM), one of the most malignant brain tumors in the world, has limited treatment options and a dismal survival rate. Effective and safe disease-modifying drugs for glioblastoma are urgently needed. Here, we identified a small molecule, Molephantin (EM-5), effectively penetrated the blood-brain barrier (BBB) and demonstrated notable antitumor effects against GBM with good safety profiles both in vitro and in vivo. Mechanistically, EM-5 not only inhibits the proliferation and invasion of GBM cells but also induces cell apoptosis through the reactive oxygen species (ROS)-mediated PI3K/Akt/mTOR pathway. Furthermore, EM-5 causes mitochondrial dysfunction and blocks mitophagy flux by impeding the fusion of mitophagosomes with lysosomes. It is noteworthy that EM-5 does not interfere with the initiation of autophagosome formation or lysosomal function. Additionally, the mitophagy flux blockage caused by EM-5 was driven by the accumulation of intracellular ROS. In vivo, EM-5 exhibited significant efficacy in suppressing tumor growth in a xenograft model. Collectively, our findings not only identified EM-5 as a promising, effective, and safe lead compound for treating GBM but also uncovered its underlying mechanisms from the perspective of apoptosis and mitophagy.


Assuntos
Apoptose , Neoplasias Encefálicas , Proliferação de Células , Glioblastoma , Mitofagia , Espécies Reativas de Oxigênio , Ensaios Antitumorais Modelo de Xenoenxerto , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Humanos , Mitofagia/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Camundongos , Proliferação de Células/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos Nus , Serina-Treonina Quinases TOR/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo
2.
PLoS Pathog ; 19(4): e1011316, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37058447

RESUMO

The presence of human cytomegalovirus (HCMV) in glioblastoma (GBM) and improved outcomes of GBM patients receiving therapies targeting the virus have implicated HCMV in GBM progression. However, a unifying mechanism that accounts for the contribution of HCMV to the malignant phenotype of GBM remains incompletely defined. Here we have identified SOX2, a marker of glioma stem cells (GSCs), as a key determinant of HCMV gene expression in gliomas. Our studies demonstrated that SOX2 downregulated promyelocytic leukemia (PML) and Sp100 and consequently facilitated viral gene expression by decreasing the amount of PML nuclear bodies in HCMV-infected glioma cells. Conversely, the expression of PML antagonized the effects of SOX2 on HCMV gene expression. Furthermore, this regulation of SOX2 on HCMV infection was demonstrated in a neurosphere assay of GSCs and in a murine xenograft model utilizing xenografts from patient-derived glioma tissue. In both cases, SOX2 overexpression facilitated the growth of neurospheres and xenografts implanted in immunodeficient mice. Lastly, the expression of SOX2 and HCMV immediate early 1 (IE1) protein could be correlated in tissues from glioma patients, and interestingly, elevated levels of SOX2 and IE1 were predictive of a worse clinical outcome. These studies argue that HCMV gene expression in gliomas is regulated by SOX2 through its regulation of PML expression and that targeting molecules in this SOX2-PML pathway could identify therapies for glioma treatment.


Assuntos
Glioma , Proteínas Imediatamente Precoces , Animais , Humanos , Camundongos , Citomegalovirus/fisiologia , Regulação para Baixo , Expressão Gênica , Glioma/genética , Glioma/patologia , Proteínas Imediatamente Precoces/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Sci Rep ; 12(1): 11181, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35778451

RESUMO

Tumor immune microenvironment exerts a profound effect on the population of infiltrating immune cells. Tissue inhibitor of matrix metalloproteinase 1 (TIMP1) is frequently overexpressed in a variety of cells, particularly during inflammation and tissue injury. However, its function in cancer and immunity remains enigmatic. In this study, we find that TIMP1 is substantially up-regulated during tumorigenesis through analyzing cancer bioinformatics databases, which is further confirmed by IHC tissue microarrays of clinical samples. The TIMP1 level is significantly increased in lymphocytes infiltrating the tumors and correlated with cancer progression, particularly in GBM. Notably, we find that the transcriptional factor Sp1 binds to the promoter of TIMP1 and triggers its expression in GBM. Together, our findings suggest that the Sp1-TIMP1 axis can be a potent biomarker for evaluating immune cell infiltration at the tumor sites and therefore, the malignant progression of GBM.


Assuntos
Glioblastoma , Linfócitos do Interstício Tumoral , Fator de Transcrição Sp1 , Inibidor Tecidual de Metaloproteinase-1 , Carcinogênese , Linhagem Celular Tumoral , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/imunologia , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/imunologia , Microambiente Tumoral/imunologia
4.
BMC Cancer ; 22(1): 499, 2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35524230

RESUMO

BACKGROUND: Atypical teratoid/rhabdoid tumor (AT/RT) is a malignant pediatric tumor of the central nervous system (CNS) with high recurrence and low survival rates that is often misdiagnosed. MicroRNAs (miRNAs) are involved in the tumorigenesis of numerous pediatric cancers, but their roles in AT/RT remain unclear. METHODS: In this study, we used miRNA sequencing and gene expression microarrays from patient tissue to study both the miRNAome and transcriptome traits of AT/RT. RESULTS: Our findings demonstrate that 5 miRNAs were up-regulated, 16 miRNAs were down-regulated, 179 mRNAs were up-regulated and 402 mRNAs were down-regulated in AT/RT. qPCR revealed that hsa-miR-17-5p and MAP7 mRNA were the most significantly differentially expressed miRNA and mRNA in AT/RT tissues. Furthermore, the results from analyses using the miRTarBase database identified MAP7 mRNA as a target gene of hsa-miR-17-5p. CONCLUSIONS: Our findings suggest that the dysregulation of hsa-miR-17-5p may be a pivotal event in AT/RT and miRNAs that may represent potential therapeutic targets and diagnostic biomarkers.


Assuntos
Neoplasias do Sistema Nervoso Central , MicroRNAs , Tumor Rabdoide , Neoplasias do Sistema Nervoso Central/genética , Criança , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro/genética , Tumor Rabdoide/genética
5.
Lab Invest ; 102(7): 731-740, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35332262

RESUMO

The WHO (2021) Classification classified a group of pediatric-type high-grade gliomas as IDH wildtype, H3 wildtype but as of currently, they are characterized only by negative molecular features of IDH and H3. We recruited 35 cases of pediatric IDH wildtype and H3 wildtype hemispheric glioblastomas. We evaluated them with genome-wide methylation profiling, targeted sequencing, RNAseq, TERT promoter sequencing, and FISH. The median survival of the cohort was 27.6 months. With Capper et al.'s36 methylation groups as a map, the cases were found to be epigenetically heterogeneous and were clustered in proximity or overlay of methylation groups PXA-like (n = 8), LGG-like (n = 10), GBM_MYCN (n = 9), GBM_midline (n = 5), and GBM_RTKIII (n = 3). Histology of the tumors in these groups was not different from regular glioblastomas. Methylation groups were not associated with OS. We were unable to identify groups specifically characterized by EGFR or PDGFRA amplification as proposed by other authors. EGFR, PDGFRA, and MYCN amplifications were not correlated with OS. 4/9 cases of the GBM_MYCN cluster did not show MYCN amplification; the group was also enriched for EGFR amplification (4/9 cases) and the two biomarkers overlapped in two cases. Overall, PDGFRA amplification was found in only four cases and they were not restricted to any groups. Cases in proximity to GBM_midline were all hemispheric and showed loss of H3K27me3 staining. Fusion genes ALK/NTRK/ROS1/MET characteristic of infantile glioblastomas were not identified in 17 cases successfully sequenced. BRAF V600E was only found in the PXA group but CDKN2A deletion could be found in other methylation groups. PXA-like cases did not show PXA histological features similar to findings by other authors. No case showed TERT promoter mutation. Mutations of mismatch repair (MMR) genes were poor prognosticators in single (p ≤ 0.001) but not in multivariate analyses (p = 0.229). MGMT had no survival significance in this cohort. Of the other common biomarkers, only TP53 and ATRX mutations were significant poor prognosticators and only TP53 mutation was significant after multivariate analyses (p = 0.024). We conclude that IDH wildtype, H3 wildtype pediatric hemispheric glioblastomas are molecularly heterogeneous and in routine practice, TP53, ATRX, and MMR status could profitably be screened for risk stratification in laboratories without ready access to methylation profiling.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/patologia , Criança , Receptores ErbB/genética , Humanos , Mutação , Proteína Proto-Oncogênica N-Myc/genética , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética
6.
Clin Neurol Neurosurg ; 208: 106882, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34428613

RESUMO

The diagnostic role of Isocitrate Dehydrogenase (IDH) mutation status in adult lower grade astrocytomas was first formally presented within the WHO Classification of Tumours of the Central Nervous System (2016). IDH-mutant astrocytomas are not as common as IDH-wildtype astrocytomas but are of better prognosis. Our previous study provided an evident that IDH-mutant lower grade astrocytomas is not a homogeneous group and could be further stratified by PDGFRA amplification, CDK4 amplification and CDKN2A deletion. In this study, we detected the expressions of DNA mismatch repair (MMR) proteins (PMS2, MLH1, MSH2, MSH6) and PD-L1 by immunohistochemistry in 147 IDH-mutant lower grade astrocytomas and explored their clinical relevance. The loss of was identified in 28.6%, 1.4%, 8.8% and 13.6%, respectively. PD-L1 expression was detected in 1.4% of this cohort. Survival analysis revealed that loss of PMS2 was correlated with shorter OS (p < 0.001) and PFS (p = 0.005). Loss of PMS2 or MLH1 was associated with shorter OS (p < 0.001) and PFS (p = 0.008). In IDH-mutant lower grade astrocytomas without CDKN2A deletion, loss of PMS2 was associated with poorer OS (p < 0.001) and PFS (p = 0.001). Furthermore, among IDH-mutant lower grade astrocytomas lacking the three biomarkers (PDGFRA, CDK4 and CDKN2A), loss of PMS2 was also associated with a poorer OS (p < 0.001) and PFS (p = 0.003). Our data illustrated the potential application of MMR genes in stratification of IDH-mutant lower grade astrocytomas without PDGFRA, CDK4 and CDKN2A copy number alterations.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Isocitrato Desidrogenase/genética , Endonuclease PMS2 de Reparo de Erro de Pareamento/genética , Proteína 1 Homóloga a MutL/genética , Adulto , Astrocitoma/metabolismo , Astrocitoma/mortalidade , Astrocitoma/patologia , Biomarcadores Tumorais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Feminino , Humanos , Isocitrato Desidrogenase/metabolismo , Masculino , Pessoa de Meia-Idade , Endonuclease PMS2 de Reparo de Erro de Pareamento/metabolismo , Proteína 1 Homóloga a MutL/metabolismo , Mutação , Prognóstico , Taxa de Sobrevida
9.
J Neurol Sci ; 414: 116837, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32334273

RESUMO

Glioblastoma multiforme (GBM) is the most common primary malignancy in the adult central nervous, and is characterized by high aggressiveness and a high mortality rate. The high mortality rate is largely due to the development of drug resistance. Temozolomide (TMZ) resistance is considered to be one of the major reasons responsible for GBM therapy failure. CXCL12/CXCR4 has been demonstrated to be involved in cell proliferation, migration, invasion, angiogenesis, and radioresistance in GBM. However, its role in TMZ resistance in GBM is unknown. In this study, we aimed to evaluate the role of CXCL12/CXCR4 in mediating the TMZ resistance to GBM cells and explore the underlying mechanisms. We found that the CXCL12/CXCR4 axis enhanced TMZ resistance in GBM cells. Further study showed that CXCL12/CXCR4 conferred TMZ resistance and promoted the migration and invasion of GBM cells by up-regulating FOXM1. This resistance was partially reversed by suppressing CXCL12/CXCR4 and FOXM1 silencing. Our study revealed the vital role of CXCL12/CXCR4 in mediating the resistance of GBM cells to TMZ, and suggested that targeting CXCL12/CXCR4 axis may attenuate the resistance to TMZ in GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Quimiocina CXCL12/genética , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Humanos , Receptores CXCR4/genética , Receptores CXCR4/uso terapêutico , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Regulação para Cima
10.
J Cell Physiol ; 235(10): 7344-7355, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32180229

RESUMO

Glioblastoma (GBM) is the most malignant primary brain tumor in adults. Due to its invasive nature, it cannot be thoroughly eliminated. WD repeat domain 12 (WDR12) processes the 32S precursor rRNA but cannot affect the synthesis of the 45S/47S primary transcript. In this study, we found that WDR12 is highly expressed in GBM according to the analysis results of mRNA expression by The Cancer Genome Atlas database. The high expression level of WDR12 is dramatically related to shorter overall survival and reduced disease-free survival. Next, we knocked down WDR12 and found that knockdown of WDR12 promoted the apoptosis and inhibited the proliferation by cell biology experiments. Differential expression genes in gene-chip revealed that WDR12 knockdown mainly inhibited cell cycle. Finally, we also found that WDR12 is associated with PLK1 and EZH2 in cell proliferation of GBM. Resumptively, this report showed a possible evidence that WDR12 drove malignant behavior of GBM, whose expression may present a neoteric independent prognostic biomarker in GBM.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/genética , Oncogenes/genética , Proteínas de Ligação a RNA/genética , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Genômica/métodos , Glioblastoma/patologia , Humanos , Prognóstico , RNA Mensageiro/genética
12.
Medicine (Baltimore) ; 99(8): e19178, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32080099

RESUMO

RATIONALE: Primary melanocytic tumors of central nerve system (CNS) are rare, primary diffuse leptomeningeal melanomatosis (PDLM), a subtype of malignant melanomas of CNS, is extremely rare,especially in pediatrics. As the clinical manifestation of PDLM is not characteristic, It is often misdiagnosed as tubercular meningitis and hemorrhage. PATIENT CONCERNS: A 13-year-old boy was admitted to our department with symptoms of recurrent headache and vomiting twice. As the brain imaging revealed a lesion located in the left temporal lobe mimicked hemorrhage, so there was a misdiagnosis of hemorrhage in first hospitalization. He was admitted again for the recurrence of the headache and vomiting. Detailed physical examination showed multiple melanin changes in the skin of the whole body which were ignored in last hospitalization. Brain imaging showed the significantly enlarged lesion in the left temporal lobe and several smaller lesions in the left parietal lobe and cerebellum which indicated metastasis. DIAGNOSIS: According to the history,physical examination and the radiological finding, the patient was diagnosed with malignant melanoma of central never system possibly. INTERVENTIONS: The patient underwent left temporal and parietal lesions total resection with a craniotomy. OUTCOMES: The diagnosis of PDLM was established according to pathological characteristics and the negative finding of positron emission tomography (PET)-computed tomography (CT) outside CNS. The patient got no further treatment for economic reasons and experienced the progression and died 5 months after operation. LESSONS: PDLM is extremely rare in CNS, as the clinical manifestation, radiological changes are not special, early diagnosis is difficult. The confirmed diagnosis is established by leptomeningeal biospy or surgical tissue. PET-CT can help differential diagnosis with metastastic leptomeningeal melanomas. The prognosis is dismal due to the inefficiency of chemotherapy or radiotherapy.


Assuntos
Melanoma/diagnóstico , Melanoma/patologia , Neoplasias Meníngeas/diagnóstico , Neoplasias Meníngeas/patologia , Adolescente , Craniotomia , Humanos , Masculino , Melanoma/diagnóstico por imagem , Melanoma/cirurgia , Neoplasias Meníngeas/diagnóstico por imagem , Neoplasias Meníngeas/cirurgia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada
13.
World Neurosurg ; 120: 177-180, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30170143

RESUMO

BACKGROUND: Cerebral cavernous malformations (CCMs) are vascular malformations that account for 5%-15% of all central nervous system vascular malformations. However, multiple CCMs, which can be sporadic or familial, are rare, with a prevalence of 0.1%-0.5%. CASE DESCRIPTION: Here, we presented a rare case of sporadic multiple CCMs in an infant, which were accompanied with multiple cavernous malformations of the chest and skin. CONCLUSIONS: CCMs were pathologically diagnosed through the total resection of the pineal regional lesion. We also observed a spontaneous regression of the remaining lesions during a follow-up period of 2 years. To our knowledge, this is the first case of CCMs in an infant in the English-language literature.


Assuntos
Neoplasias Encefálicas/diagnóstico , Hemangioma Cavernoso do Sistema Nervoso Central/diagnóstico , Neoplasias Primárias Múltiplas/diagnóstico , Neoplasias Cutâneas/diagnóstico , Neoplasias Torácicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/cirurgia , Derivações do Líquido Cefalorraquidiano , Feminino , Hemangioma Cavernoso do Sistema Nervoso Central/genética , Hemangioma Cavernoso do Sistema Nervoso Central/cirurgia , Humanos , Hidrocefalia/diagnóstico , Hidrocefalia/genética , Hidrocefalia/cirurgia , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Regressão Neoplásica Espontânea , Neoplasias Primárias Múltiplas/genética , Neoplasias Primárias Múltiplas/cirurgia , Pinealoma/diagnóstico , Pinealoma/genética , Pinealoma/cirurgia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Gravidez , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/cirurgia , Neoplasias Torácicas/genética , Tomografia Computadorizada por Raios X , Ultrassonografia Pré-Natal
14.
Int J Oncol ; 53(5): 2067-2080, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30106127

RESUMO

Glioblastoma multiforme (GBM) is the most common primary malignant tumor affecting the human brain. Despite improvements in therapeutic technologies, patients with GBM have a poor clinical result and the molecular mechanisms responsible for the development of GBM have not yet been fully elucidated. 3-phosphoinositide dependent protein kinase 1 (PDK1) is upregulated in various tumors and promotes tumor invasion. In glioma, transforming growth factor-ß (TGF­ß) promotes cell invasion; however, whether TGF­ß directly regulates PDK1 protein and promotes proliferation and invasion is not yet clear. In this study, PDK1 levels were measured in glioma tissues using tissue microarray (TMA) by immunohistochemistry (IHC) and RT­qPCR. Kaplan-Meier analyses were used to calculate the survival rate of patients with glioma. In vitro, U251 and U87 glioma cell lines were used for functional analyses. Cell proliferation and invasion were analyzed using siRNA transfection, MTT assay, RT­qPCR, western blot analysis, flow cytometry and invasion assay. In vivo, U251 glioma cell xenografts were established. The results revealed that PDK1 protein was significantly upregulated in glioma tissues compared with non-tumorous tissues. Furthermore, the higher PDK1 levels were associated with a large tumor size (>5.0 cm), a higher WHO grade and a shorter survival of patients with GBM. Univariate and multivariate analyses indicated that PDK1 was an independent prognostic factor. In vivo, PDK1 promoted glioma tumor xenograft growth. In vitro, functional analyses confirmed that TGF­ß upregulated PDK1 protein expression and PDK1 promoted cell migration and invasion, and functioned as an oncogene in GBM, by upregulating c­Jun protein and inducing epithelial-mesenchymal transition (EMT). c­Jun protein were overexpressed in glioma tissues and positively correlated with PDK1 levels. Moreover, our findings were further validated by the online Oncomine database. On the whole, the findings of this study indicate that in GBM, PDK1 functions as an oncogene, promoting proliferation and invasion.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Neoplasias Encefálicas/patologia , Glioma/patologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/genética , Adulto , Idoso , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Glioblastoma/patologia , Glioma/metabolismo , Glioma/mortalidade , Humanos , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Oncogenes , Proteínas Proto-Oncogênicas c-jun/genética , Taxa de Sobrevida , Fator de Crescimento Transformador beta/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Int J Mol Med ; 41(4): 2288-2296, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29344634

RESUMO

Photodynamic therapy (PDT) is a relatively novel type of tumor therapy method with low toxicity and limited side­effects. The aim of the present study was to investigate the underlying mechanism and potential microRNAs (miRNAs) involved in the treatment of glioma by PDT with hematoporphyrin, a clinical photosensitizer. The photodynamic activity of hematoporphyrin on the cell viability and apoptosis of gliomas was investigated by MTT, and flow cytometry and fluorescence microscopy, respectively. Alterations in singlet oxygen and mitochondrial membrane potential were detected. The differentially expressed miRNAs and proteins were evaluated by miRNA gene chip and apoptosis­associated protein chip, respectively. The results demonstrated that cell viability significantly decreased with hematoporphyrin concentration. PDT with hematoporphyrin significantly increased cell apoptosis at a later stage, induced the content of reactive oxygen species (ROS) and decreased the mitochondrial membrane potential, indicating that PDT with hematoporphyrin inhibited cell growth via induction of radical oxygen, decreased the mitochondrial membrane potential and induced apoptosis. The upregulated miRNAs, including hsa­miR­7641, hsa­miR­9500, hsa­miR­4459, hsa­miR­21­5p, hsa­miR­663a and hsa­miR­205­5p may be important in PDT­induced cell apoptosis in glioma. Transporter 1, ATP binding cassette subfamily B member­ and nuclear factor­κB­mediated apoptosis signaling pathways were the most significant pathways. Thus, the current study presents PDT as a potential therapeutic approach for the treatment of malignant glioma, and identified miRNAs for the molecular design and development of a third­generation photosensitizer (PS).


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Hematoporfirinas/farmacologia , MicroRNAs/genética , Fármacos Fotossensibilizantes/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Fotoquimioterapia , Oxigênio Singlete/metabolismo
16.
Int J Oncol ; 52(2): 485-495, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29345288

RESUMO

Glioblastoma (GBM) is the most common type of malignant brain tumor. In spite of recent advancements in surgical techniques, chemotherapy, and radiation therapy, patients with GBM often face a dire prognosis. MicroRNAs have been shown to modulate the aggressiveness of various cancers, and have emerged as possible therapeutic agents for the management of GBM. miR­205 is dysregulated in glioma and act as a prognostic indicator. However, the role of miR­205 in the development of GBM has not been elucidated. To better understand the pathogenesis of GBM, we examine the biological significance and molecular mechanisms of miR­205 in GBM cells. Zinc finger E-box binding homeobox 1 (ZEB1) has been shown to regulate the epithelial-mesenchymal transition (EMT), which is strongly associated with GBM malignancy. In the present study, we show miR­205 expression is reduced in GBM tissues and cell lines, and ZEB1 expression is inversely correlated with miR­205 expression. We also show ZEB1 is a downstream target of miR­205 and the Akt/mTOR signaling pathway is activated when miR­205 interacts with ZEB1. Increased activity of miR­205 in GBM cells significantly inhibits migration and invasion, and prevents EMT. Furthermore, overexpression of ZEB1 partially abolishes these inhibitory effects of miR­205. We show that miR­205 negatively regulates the expression of ZEB1 in GBM, inhibits cell migration and invasion, and prevents EMT, at least in part through the inhibition of the activation of the Akt/mTOR signaling pathway. Our results indicate miR­205 may be an efficacious therapeutic agent in the treatment of GBM.


Assuntos
Neoplasias Encefálicas/patologia , Transição Epitelial-Mesenquimal/genética , Glioblastoma/patologia , MicroRNAs/genética , Regiões 3' não Traduzidas , Adulto , Idoso , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
17.
Cardiovasc Ther ; 36(1)2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29171187

RESUMO

BACKGROUND: The development of heart failure after acute myocardial infarction (MI) was related to left ventricular (LV) pathological remodeling and dysfunction. Cardiac stem cells (CSCs) provided a new option to treat acute MI. This study was to investigate the effects of CSCs on structural and functional alteration in acute MI. METHODS: Acute MI was induced in 20 Sprague-Dawley rats by ligation of the left anterior descending coronary artery. Two weeks after MI, animals were randomized into CSCs or control group. LV geometry and function were echocardiographically measured at baseline, 2, 4, and 6 weeks after MI. After measuring hemodynamics at 6 weeks after MI, hearts were harvested for tracing CSCs stained by PKH26 and testing expression of VEGF-α/TGF-ß1 by RT-PCR and ELISA. RESULTS: Two weeks after MI, there were significant decreases in interventricular septal systolic and diastolic thickness (IVSTs/d), while increases in LV systolic and diastolic dimension (LVDs/d). Consequently, this contributed to decreases in ejection fraction (EF) and fractional shorting (FS). With the treatment of CSCs for 4 weeks, significant better ejection fraction (EF) and fractional shorting (FS) were achieved in CSCs group accompanied by the reduction in LV systole and diastole dimension (LVDs/d). Besides, a significant improvement in the maximal rate of LV pressure development and decline (peak +dP/dt and -dP/dt, respectively) was observed. Moreover, significantly higher VEGF-α was expressed in CSCs group rather than TGF-ß1. CONCLUSION: CSCs significantly prevented structural and functional deterioration after MI with increasing expression of VEGF-α.


Assuntos
Infarto do Miocárdio/cirurgia , Miocárdio , Transplante de Células-Tronco , Células-Tronco , Disfunção Ventricular Esquerda/prevenção & controle , Função Ventricular Esquerda , Remodelação Ventricular , Animais , Células Cultivadas , Modelos Animais de Doenças , Masculino , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Fenótipo , Ratos Sprague-Dawley , Regeneração , Células-Tronco/metabolismo , Volume Sistólico , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo , Transplante Homólogo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia , Pressão Ventricular
18.
Onco Targets Ther ; 10: 2173-2181, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28458560

RESUMO

Glioma is the most common and aggressive human primary tumor in the central nervous system. Despite present clinical advancements, median survival time remains poor in this malignant tumor. Serpin peptidase inhibitor, clade A member 3 (SERPINA3), is a member of the serpin superfamily of protease inhibitors. Its aberrant expression has been observed in various tumors. However, its clinical significance and biological function in glioma remain unclear, especially for the prognosis of glioma patients. In this study, we investigated SERPINA3 expression in glioma tissue samples and its significance in predicting the prognosis of glioma patients. SERPINA3 protein expression was studied by immunohistochemistry, while real-time polymerase chain reaction was used to study SERPINA3 mRNA expression. We found that SERPINA3 was upregulated in glioma tissue at both mRNA and protein levels, compared with noncancerous brain tissues. We also found that high SERPINA3 expression in glioma tissues correlated significantly with advanced World Health Organization grade. Univariate and multivariate analyses revealed that high SERPINA3 expression was an independent prognostic factor for poor overall survival of glioma patients. Moreover, our findings were further validated by online Oncomine database. Taken together, our results suggest that SERPINA3 plays an oncogenic role in glioma progression and provide an insight into the application of SERPINA3 as a novel predictor of clinical outcomes and a potential biomarker of glioma.

19.
Oncotarget ; 8(14): 22783-22799, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28187000

RESUMO

Glioblastoma multiforme (GBM) is the most malignant brain tumor with limited therapeutic options. Temozolomide (TMZ) is a novel cytotoxic agent used as first-line chemotherapy for GBM, however, some individual cells can't be isolated for surgical resection and show treatment-resistance, thus inducing poor prognosis. By using the HiSeq sequencing and bioinformatics methods, we identified lncRNAs showing different expression levels in TMZ-resistant and non-resistant patients. RT-qPCR was then performed in tissues and serum samples, and lncRNA MALAT1 was finally identified to show considerable discriminating potential to identify responding patients from non-responding patients. Moreover, high serum MALAT1 expression was associated with poor chemoresponse and survival in GBM patients receiving TMZ treatment. Subsequently, the TMZ resistant cell lines were established, and the CCK8 assay showed that lncRNA MALAT1 knockdown significantly reversed TMZ resistance in GBM cells. The gain and loss-function experiments revealed that miR-203 was down-regulated by MALAT1 and this interaction has reciprocal effects. Besides, thymidylate synthase (TS) mRNA was identified as a direct target of miR-203. LncRNA MALAT1 inhibition re-sensitized TMZ resistant cells through up-regulating miR-203 and down-regulating TS expression. On the other hand, MALAT1 overexpression promoted resistance by suppressing miR-203 and promoting TS expression. In conclusion, our integrated approach demonstrates that enhanced expression of lncRNA MALAT1 confers a potent poor therapeutic efficacy and inhibition of MALAT1 levels could be a future direction to develop a novel therapeutic strategy to overcome TMZ resistance in GBM patients.


Assuntos
Neoplasias Encefálicas/patologia , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/patologia , MicroRNAs/genética , RNA Longo não Codificante/genética , Timidilato Sintase/metabolismo , Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dacarbazina/farmacologia , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Estadiamento de Neoplasias , Prognóstico , Taxa de Sobrevida , Temozolomida , Timidilato Sintase/genética , Células Tumorais Cultivadas
20.
Tumour Biol ; 37(4): 5075-87, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26546435

RESUMO

Cytoplasmic polyadenylation element binding protein 4 (CPEB4) is a regulator of gene transcription and has been reported to be associated with biological malignancy in cancers. However, it is unclear whether CPEB4 has any clinical significance in patients with astrocytic tumors, and mechanisms that CPEB4 contribute to progression of astrocytic tumors remain largely unknown. Here, correlation between CPEB4 expression and prognosis of patients with astrocytic tumors were explored by using qPCR, WB and IHC, and X-tile, SPSS software. Cell lines U251 MG and A172 were used to study CPEB4's function and mechanisms. Co-immunoprecipitation, mass spectrometry, immunofluorescent assay, and western blot were performed to observe the interaction between CPEB4 and Vimentin. CPEB4 mRNA and protein levels were markedly elevated in 12/12 astrocytic tumors in comparison to paratumor. High expression of CPEB4 was significantly correlated with clinical progressive futures and work as an independent adverse prognostic factor for overall survival of patients with astrocytic tumors (relative risk 4.5, 95 % CI 2.1-11.2, p = 0.001). Moreover, knockdown of CPEB4 in astrocytic tumor cells inhibited their proliferation ability , clonogenicity, and invasiveness. Five candidate proteins, GRP78, Mortalin, Keratin, Vimentin, and ß-actin, were identified, and the interaction between CPEB4 and Vimentin was finally confirmed. Downregulation of CPEB4 could reduce the protein expression of Vimentin. Our studies first validated that CPEB4 interacts with Vimentin and indicated that high CPEB4 expression in astrocytic tumors correlates closely with a clinically aggressive future, and that CPEB4 might represent a valuable prognostic marker for patients with astrocytic tumors.


Assuntos
Astrocitoma/genética , Biomarcadores Tumorais/genética , Prognóstico , Proteínas de Ligação a RNA/genética , Vimentina/genética , Actinas/genética , Adulto , Idoso , Astrocitoma/patologia , Astrocitoma/cirurgia , Biomarcadores Tumorais/biossíntese , Proliferação de Células/genética , Chaperona BiP do Retículo Endoplasmático , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico/biossíntese , Humanos , Queratinas/biossíntese , Masculino , Pessoa de Meia-Idade , Proteínas Mitocondriais/biossíntese , Invasividade Neoplásica/genética , RNA Mensageiro/biossíntese , Proteínas de Ligação a RNA/biossíntese , Análise de Sobrevida , Vimentina/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA