Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Eur J Pharmacol ; 894: 173853, 2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33422507

RESUMO

Acute promyelocytic leukemia (APL) is associated with PML-RARα oncogene, which is treated using all-trans retinoic acid (ATRA)-based chemotherapy. However, chemoresistance is observed in 20-30% of treated patients and represents a clinical challenge, raising the importance of the development of new therapeutic options. In the present study, the effects of three synthetic cyclopenta[b]indoles on the leukemia phenotype were investigated using NB4 (ATRA-sensitive) and NB4-R2 (ATRA-resistant) cells. Among the tested synthetic cyclopenta[b]indoles, compound 2, which contains a heterocyclic nucleus, was the most active, presenting time-dependent cytotoxic activity in the µM range in APL cells, without cytotoxicity for normal leukocytes, and was selected for further characterization. Compound 2 significantly decreased clonogenicity, increased apoptosis, and caused cell cycle arrest at S and G2/M phases in a drug concentration-dependent manner. Morphological analyses indicated aberrant mitosis and diffuse tubulin staining upon compound 2 exposure, which corroborates cell cycle findings. In the molecular scenario, compound 2 reduced STMN1 expression and activity, and induced PARP1 cleavage and H2AX and CHK2 phosphorylation, and modulated CDKN1A, PMAIP1, GADD45A, and XRCC3 expressions, indicating reduction of cell proliferation, apoptosis, and DNA damage. Moreover, in the in vivo tubulin polymerization assay, NB4 and NB4-R2 cells showed a reduction in the levels of polymerized tubulin upon compound 2 exposure, which indicates tubulin as a target of the drug. Molecular docking supports this hypothesis. Taken together, these data indicated that compound 2 exhibits antileukemic effects through disrupting the microtubule dynamics, identifying a possible novel potential antineoplastic agent for the treatment of ATRA-resistant APL.


Assuntos
Antineoplásicos/farmacologia , Ciclopentanos/química , Indóis/farmacologia , Leucemia Promielocítica Aguda/tratamento farmacológico , Microtúbulos/metabolismo , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Indóis/química , Microtúbulos/efeitos dos fármacos , Mitose/efeitos dos fármacos , Modelos Moleculares , Estatmina/biossíntese , Ensaio Tumoral de Célula-Tronco
2.
Drug Des Devel Ther ; 13: 2067-2079, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31308624

RESUMO

Background: Understanding of lidocaine-induced neurotoxicity is not complete, resulting in the unsuccessful treatment in some clinical settings. Dexmedetomidine (DEX) has been shown to alleviate lidocaine-induced neurotoxicity in our previous cell model. However, the rationale for DEX combined with lidocaine to reduce lidocaine-induced neurotoxicity in the clinical setting remains to be further clarified in the detailed molecular mechanism. Methods: In this study, we established a cellular injury model by lidocaine preconditioning. Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) proliferation assay kit were used to analyze cell proliferation. Cell apoptosis was measured by flow cytometry and Hoechst 33342 staining. Cell cycle progression was detected by flow cytometry. The protein expression levels were detected by Western blotting and immunofluorescence staining. Results: Our results showed that DEX dose-dependently restored impaired proliferation of PC12 cells induced by lidocaine,as reflected by the increased cell viability and EdU positive cells, which were consistent with the decreased expression of tumor suppressor protein p21 and increased expression of cell cycle-related cyclin D1 and CDK1. In addition, DEX dose-dependently reduced apoptotic PC12 cells induced by lidocaine,as reflected by the decreased expression of apoptosis-related Bax, caspase-3 and caspase-9 and increased expression of anti-apoptotic Bcl-2 compared to the cells only treated with lidocaine. Mechanistically, with gain-or-loss-of-function of STMN1, we showed that DEX-mediated neuroprotection by lidocaine-induced damage is associated with downregulation of STMN1 which might be an upstream molecule involved in regulation of mitochondria death pathway. Conclusion: Our results reveal that DEX is likely to be an effective adjunct to alleviate chronic neurotoxicity induced by lidocaine.


Assuntos
Dexmedetomidina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Lidocaína/farmacologia , Substâncias Protetoras/farmacologia , Estatmina/biossíntese , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Lidocaína/antagonistas & inibidores , Células PC12 , Ratos
3.
J Cell Biochem ; 119(5): 4009-4020, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29231257

RESUMO

Traditional Chinese medicines have been recognized as especially promising anticancer agents in modern anticancer research. Halofuginone (HF), an analog of quinazolinone alkaloid extracted from Dichroa febrifuga, is widely used in traditional medicine. However, whether HF inhibits the growth of breast cancer cells and/or reduces the migration and invasion of MCF-7 human breast cancer cells, as well as the underlying mechanisms in vitro, remains unclear. In this study, we report that an HF extract inhibits the growth of MCF-7 cells and reduces their migration and invasion, an important feature of potential anticancer agents. In addition, HF significantly increases the activation of autophagy, which is closely associated with tumor metastasis. As STMN1 and p53 have been closely implicated in breast cancer progression, we analyzed their expression in the context of HF extract treatment. Western blot analysis showed that HF suppresses STMN1 and p53 expression and activity in an autophagy-dependent manner. Collectively, these data indicate that activation of autophagy reduces expression of STMN1 and p53, and the migration and invasion of cancer cells contributes to the anti-cancer effects of the HF. These findings may provide new insight into breast cancer prevention and therapy.


Assuntos
Autofagia/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Piperidinas/farmacologia , Quinazolinonas/farmacologia , Estatmina/biossíntese , Proteína Supressora de Tumor p53/biossíntese , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Feminino , Humanos , Células MCF-7 , Invasividade Neoplásica , Piperidinas/química , Quinazolinonas/química
4.
Neurosci Lett ; 654: 1-5, 2017 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-28625575

RESUMO

There is a small part of the pathological type of the meningioma has a malignant tendency and patients have the poor prognosis. Looking for effective biomarkers to predict the degree of malignancy of the tumors, will help us to better manage the patient and guide the treatment. The present study aims at investigating the prognostic value of the expression of Stathmin in a series of meningiomas of different grade. We integrated eight published microarray datasets of meningiomas to screen grade biomarkers in meningiomas patients using the WebArrayDB platform. We focused on Stathmin, Using formalin-fixed paraffin-embedded (FFPE) tumor samples, we corroborated the relationship between Stathmin and patient outcomes using qRT-PCR for gene expression. We also found expression of Stathmin that atypical/anaplastic meningiomas have higher expression than benign meningiomas (p<0.01). No correlation between Stathmin expression and age, gender and tumor extent of resection was found (p>0.05). Moreover, increased Stathmin expression was correlated to higher meningioma grade and shorter disease-free survival (DFS) of meningioma patients with Simpson I resection. Stathmin might be promising targets to improve the cure rates in meningiomas.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Meníngeas/patologia , Meningioma/patologia , Estatmina/biossíntese , Adolescente , Adulto , Idoso , Intervalo Livre de Doença , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Neoplasias Meníngeas/mortalidade , Meningioma/mortalidade , Pessoa de Meia-Idade , Prognóstico , Estatmina/análise , Regulação para Cima , Adulto Jovem
5.
Biochim Biophys Acta Proteins Proteom ; 1865(5): 488-498, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28216224

RESUMO

Breast cancer is the most common and molecularly relatively well characterized malignant disease in women, however, its progression to metastatic cancer remains lethal for 78% of patients 5years after diagnosis. Novel markers could identify the high risk patients and their verification using quantitative methods is essential to overcome genetic, inter-tumor and intra-tumor variability and translate novel findings into cancer diagnosis and treatment. We recently identified 13 proteins associated with estrogen receptor, tumor grade and lymph node status, the key factors of breast cancer aggressiveness, using untargeted proteomics. Here we verified these findings in the same set of 96 tumors using targeted proteomics based on selected reaction monitoring with mTRAQ labeling (mTRAQ-SRM), transcriptomics and immunohistochemistry and validated in 5 independent sets of 715 patients using transcriptomics. We confirmed: (i) positive association of anterior gradient protein 2 homolog (AGR2) and periostin (POSTN) and negative association of annexin A1 (ANXA1) with estrogen receptor status; (ii) positive association of stathmin (STMN1), cofilin-1 (COF1), plasminogen activator inhibitor 1 RNA-binding protein (PAIRBP1) and negative associations of thrombospondin-2 (TSP2) and POSTN levels with tumor grade; and (iii) positive association of POSTN, alpha-actinin-4 (ACTN4) and STMN1 with lymph node status. This study highlights a panel of gene products that can contribute to breast cancer aggressiveness and metastasis, the understanding of which is important for development of more precise breast cancer treatment.


Assuntos
Fatores de Despolimerização de Actina/biossíntese , Neoplasias da Mama/genética , Moléculas de Adesão Celular/biossíntese , Proteínas de Ligação a RNA/biossíntese , Estatmina/biossíntese , Trombospondinas/biossíntese , Fatores de Despolimerização de Actina/genética , Adulto , Idoso , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Neoplasias da Mama/patologia , Moléculas de Adesão Celular/genética , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Linfonodos/metabolismo , Linfonodos/patologia , Metástase Linfática , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Prognóstico , Proteômica , Proteínas de Ligação a RNA/genética , Receptores de Estrogênio/genética , Estatmina/genética , Trombospondinas/genética
6.
Oncogene ; 36(4): 501-511, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-27321182

RESUMO

Neuroblastoma, the most common solid tumor of young children, frequently presents with aggressive metastatic disease and for these children the 5-year survival rates are dismal. Metastasis, the movement of cancer cells from one site to another, involves remodeling of the cytoskeleton including altered microtubule dynamics. The microtubule-destabilizing protein, stathmin, has recently been shown to mediate neuroblastoma metastasis although precise functions remain poorly defined. In this study we investigated stathmin's contribution to the metastatic process and potential mechanism(s) by which it exerts these effects. Stathmin suppression significantly reduced neuroblastoma cell invasion of 3D tumor spheroids into an extracellular matrix. Moreover, inhibiting stathmin expression significantly reduced transendothelial migration in two different neuroblastoma cell lines in vitro. Inhibition of ROCK, a key regulator of cell migration, in neuroblastoma cells highlighted that stathmin regulates transendothelial migration through ROCK signaling. Reduced stathmin expression in neuroblastoma cells significantly increased the activation of the RhoA small GTPase. Notably, re-expression of either wild type or a phospho-mimetic stathmin mutant (4E) made defective in tubulin binding returned cell migration and transendothelial migration back to control levels, indicating that stathmin may influence these processes in neuroblastoma cells independent of tubulin binding. Finally, stathmin suppression in neuroblastoma cells significantly reduced whole body, lung, kidney and liver metastases in an experimental metastases mouse model. In conclusion, stathmin suppression interferes with the metastatic process via RhoA/ROCK signaling in neuroblastoma cells. These findings highlight the importance of stathmin to the metastatic process and its potential as a therapeutic target for the treatment of neuroblastoma.


Assuntos
Neuroblastoma/patologia , Estatmina/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos SCID , Metástase Neoplásica , Neuroblastoma/metabolismo , Transdução de Sinais , Estatmina/biossíntese , Migração Transendotelial e Transepitelial , Transfecção , Tubulina (Proteína)/metabolismo
7.
Int J Oncol ; 50(1): 31-40, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27878293

RESUMO

Stathmin1, a microtubule-destabilizing phosphoprotein, is considered to play a crucial role in regulating cellular microtubule dynamics and controlling mitosis. Previous studies have showed that STMN1 is highly expressed in many human malignancies and is related to development, invasion and metastasis of tumors. However, its expression pattern, clinical performance and functional roles in hypopharyngeal squamous cell carcinoma (HSCC) have not been addressed. In this study, we found that STMN1 was significantly elevated in HSCC and its expression level was correlated with poor differentiation (P<0.001), clinical stage (P<0.001), large tumor size (P=0.001) and lymph node metastasis (P=0.008). A positive correlation between STMN1 and Ki­67 expression was also exhibited. High STMN1 expression predicted poor survival. Furthermore, we found that knockdown of STMN1 by siRNAs inhibited the FaDu cell proliferation and migration. Moreover, decreased STMN1 expression in FaDu cells reversed the acquisition of EMT phenotype by upregulating E-cadherin, as well as reduced vimentin expression at protein and mRNA levels. These results suggested that STMN1 plays an important role in proliferation and migration of HSCC and may be used as a potential prognostic biomarker or therapeutic target of HSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Proliferação de Células/genética , Neoplasias Hipofaríngeas/genética , Estatmina/biossíntese , Adulto , Idoso , Apoptose/genética , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Caderinas/biossíntese , Caderinas/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hipofaríngeas/patologia , Antígeno Ki-67/biossíntese , Antígeno Ki-67/genética , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Estatmina/genética
8.
Sci Rep ; 6: 28833, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27349455

RESUMO

Gallbladder carcinoma (GBC) is a highly lethal malignancy of the gastrointestinal tract. Despite extensive research, the underlying molecular mechanism of GBC remains largely unclear. Stathmin 1 (STMN1) is an important cytosolic protein associated with microtubule stability that was reported to be involved in tumorigenesis. Up to our knowledge, its role in gallbladder carcinoma has not been analyzed. In this study, we found that STMN1 was significantly highly expressed in GBC by immunohistochemistry (IHC). Further research demonstrated that silencing of STMN1 inhibited cell growth in vitro. Moreover, knockdown of STMN1 induced apoptosis and delayed G2/M phase transformation in GBC cells. Our data support a rationale for further studies that the silencing of STMN1 may regulate the activity of p38 MAPK kinase and p53/p21 signal pathway. Besides, xenografted gallbladder carcinoma cells growth were significantly impaired after STMN1 was silenced in vivo. These results suggested that STMN1 played an important role in cell proliferation and migration. This provided a potential clue for investigating the therapeutic target in GBC.


Assuntos
Proliferação de Células/genética , Neoplasias da Vesícula Biliar/genética , Interferência de RNA , Estatmina/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação para Baixo , Feminino , Neoplasias da Vesícula Biliar/metabolismo , Neoplasias da Vesícula Biliar/terapia , Humanos , Imuno-Histoquímica , Camundongos Endogâmicos BALB C , Terapêutica com RNAi/métodos , Estatmina/biossíntese , Carga Tumoral/genética
9.
Pathol Res Pract ; 211(11): 816-23, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26235036

RESUMO

Stathmin 1 (STMN1) is an important molecule in regulating cellular microtubule dynamics and promoting microtubule depolymerization in interphase and late mitosis. Evidences showed that STMN1 was up-regulated in many cancers, but there was no report about the roles of STMN1 in human cutaneous squamous cell carcinoma (cSCC). Here, we confirmed significant upregulation of STMN1 in cSCC tissues and cell lines compared with non-tumor counterparts. STMN1 upregulation was associated with the proliferation, migration, invasion and apoptosis of cSCC cells. The results suggested that STMN1 may play an important role in the development and tumor progression of cSCC.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células Escamosas/patologia , Neoplasias Cutâneas/patologia , Estatmina/biossíntese , Western Blotting , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Imuno-Histoquímica , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Estatmina/análise , Transfecção , Regulação para Cima
10.
Gynecol Oncol ; 139(1): 104-11, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26206555

RESUMO

OBJECTIVE: To credential Stathmin 1 (STMN1) and p16(INK4A) (p16) as adjunct markers for the diagnosis of serous tubal intraepithelial carcinoma (STIC), and to compare STMN1 and p16 expression in p53-positive and p53-negative STIC and invasive high-grade serous carcinoma (HGSC). METHODS: Immunohistochemistry (IHC) was used to examine STMN1 and p16 expression in fallopian tube specimens (n=31) containing p53-positive and p53-negative STICs, invasive HGSCs, and morphologically normal FTE (fallopian tube epithelium). STMN1 and p16 expression was scored semiquantitatively by four individuals. The semiquantitative scores were dichotomized, and reported as positive or negative. Pooled siRNA was used to knockdown p53 in a panel of cell lines derived from immortalized FTE and HGSC. RESULTS: STMN1 and p16 were expressed in the majority of p53-positive and p53-negative STICs and concomitant invasive HGSCs, but only scattered positive cells were present in morphologically normal FTE. Both proteins were expressed consistently across multiple STICs from the same patient and in concomitant invasive HGSC. Knockdown of p53 in immortalized FTE cells and in four HGSC-derived cell lines expressing different missense p53 mutations did not affect STMN1 protein levels. CONCLUSIONS: This study demonstrates that STMN1 and p16 are sensitive and specific adjunct biomarkers that, when used with p53 and Ki-67, improve the diagnostic accuracy of STIC. The addition of STMN1 and p16 helps to compensate for practical limitations of p53 and Ki-67 that complicate the diagnosis in up to one third of STICs.


Assuntos
Biomarcadores Tumorais/biossíntese , Cistadenocarcinoma Seroso/metabolismo , Neoplasias das Tubas Uterinas/metabolismo , Proteínas de Neoplasias/biossíntese , Neoplasias Ovarianas/metabolismo , Estatmina/biossíntese , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Neoplasias das Tubas Uterinas/genética , Neoplasias das Tubas Uterinas/patologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Mutação , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
11.
Oncotarget ; 6(26): 22227-38, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26087399

RESUMO

Currently, Stathmin1 (STMN1) and phospho-STMN1 levels in breast cancers and their clinical implications are unknown. We examined the expression of STMN1 and its serine phospho-site (Ser16, Ser25, Ser38, and Ser63) status by immunohistochemistry. Using Cox regression analysis, a STMN1 expression signature and phosphorylation profile plus clinicopathological characteristics (STMN1-E/P/C) was developed in the training set (n = 204) and applied to the validation set (n = 106). This tool enabled us to separate breast cancer patients into high- and low-risk groups with significantly different disease-free survival (DFS) rates (P < 0.001). Importantly, this STMN1-E/P/C model had a greater prognostic value than the traditional TNM classifier, especially in luminal subtype breast cancer (P = 0.002). Further analysis showed that patients in the low-risk group would benefit more from adjuvant paclitaxel-based chemotherapy (P = 0.002). In conclusion, the STMN1-E/P/C signature is a reliable prognostic indicator for luminal subtype breast cancer and may predict the therapeutic response to paclitaxel-based treatments, potentially facilitating individualized management.


Assuntos
Neoplasias da Mama/metabolismo , Estatmina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/mortalidade , Quimioterapia Adjuvante , Estudos de Coortes , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Paclitaxel/administração & dosagem , Fosforilação , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Estatmina/biossíntese , Resultado do Tratamento
12.
Tumour Biol ; 36(9): 7195-204, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25894372

RESUMO

Paclitaxel can exert therapeutic effects by interacting with microtubules. Stathmin and ß-III-tubulin, which have impact on microtubule activity, are believed to be involved in the chemotherapy. The purpose of the present study was to evaluate the associations between stathmin and ß-III-tubulin expression and treatment response and survivals in patients with non-small cell lung cancer (NSCLC). Two hundred thirty-eight patients who were treated by platinum-based chemotherapy were enrolled in this study, among them, 111 patients also received paclitaxel treatment. Formalin-fixed and paraffin-embedded tumor tissues were collected for messenger RNA (mRNA) and protein detection. We assessed the associations of the two molecules with treatment response and survival outcome. High level of stathmin exhibited poor response to chemotherapy (for mRNA, P = 0.041; for protein, P = 0.017). Overexpression of stathmin was associated with shorter overall survival (for mRNA, P = 0.012; for protein, P = 0.014) and progression-free survival (for mRNA, P = 0.039; for protein, P = 0.022). Of note, this association was only observed in patients who were treated by both platinum and paclitaxel. Similar effects were not observed for ß-III-tubulin. The findings demonstrated that paclitaxel effect may be interfered with stathmin; overexpression of stathmin is a predictive marker for a worse prognosis in patients with NSCLC who were treated by both platinum and paclitaxel chemotherapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Paclitaxel/administração & dosagem , Estatmina/biossíntese , Tubulina (Proteína)/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Intervalo Livre de Doença , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estatmina/genética , Tubulina (Proteína)/genética
13.
Cancer Biother Radiopharm ; 29(9): 376-86, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25379611

RESUMO

PURPOSE: Taxol is an effective chemotherapeutic agent against epithelial-derived carcinomas, and resistance of carcinoma cells to taxol has developed with the wide prescription of the drug. In this study, five different epithelial carcinoma cell lines were randomly employed to screen the resistant cell line to taxol, and to explore the probable mechanism of taxol-resistant development. MATERIALS AND METHODS: Cells were grouped into the controls and the taxol treated. The treatment effects of five different epithelial carcinoma cell lines, including CNE1, Hep3B-2, MGC, MCF-7, and NCI-H1299, after being treated by taxol were analyzed through inspecting the ratios of cellular apoptosis, inhibition of cellular proliferation, the capability of cell colony formation and wound recovery, and the interference of cell motility and invasion, while western blot analysis and siRNA targeting Op18/stathmin were applied to explore the probable mechanism on the taxol resistance difference in these cells. RESULTS: Nonsmall cell lung cancer NCI-H1299 cells presented obvious taxol resistance, and the inhibition of cell motility and invasion was also the weakest in taxol-treated NCI-H1299 cells among these five cell lines. Microtubule dynamics analysis demonstrated that taxol treatment destroyed normal microtubule arrays and caused obvious microtubule collapse in CNE1, Hep3B-2, MGC, and MCF-7 rather than NCI-H1299, while the latter expressed high levels of microtubule-destabilizing protein Op18/stathmin. Inhibition of Op18/stathmin expression increased the sensitivity to taxol and promoted cellular apoptosis in NCI-H1299 cells. CONCLUSION: NCI-H1299 cells are evidently resistant to taxol-induced cellular apoptosis, inhibition of cellular proliferation and wound recovery, as well as cell migration and invasion interference, which are closely associated with the changes of microtubule dynamics. High expression of Op18/stathmin is perhaps a crucial determinant of taxol-resistant development in NCI-H1299 cells.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma/tratamento farmacológico , Paclitaxel/farmacologia , Estatmina/biossíntese , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Células MCF-7 , Estatmina/genética , Transfecção
14.
PLoS Genet ; 10(8): e1004558, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25122120

RESUMO

Cytokinesis terminates mitosis, resulting in separation of the two sister cells. Septins, a conserved family of GTP-binding cytoskeletal proteins, are an absolute requirement for cytokinesis in budding yeast. We demonstrate that septin-dependence of mammalian cytokinesis differs greatly between cell types: genetic loss of the pivotal septin subunit SEPT7 in vivo reveals that septins are indispensable for cytokinesis in fibroblasts, but expendable in cells of the hematopoietic system. SEPT7-deficient mouse embryos fail to gastrulate, and septin-deficient fibroblasts exhibit pleiotropic defects in the major cytokinetic machinery, including hyperacetylation/stabilization of microtubules and stalled midbody abscission, leading to constitutive multinucleation. We identified the microtubule depolymerizing protein stathmin as a key molecule aiding in septin-independent cytokinesis, demonstrated that stathmin supplementation is sufficient to override cytokinesis failure in SEPT7-null fibroblasts, and that knockdown of stathmin makes proliferation of a hematopoietic cell line sensitive to the septin inhibitor forchlorfenuron. Identification of septin-independent cytokinesis in the hematopoietic system could serve as a key to identify solid tumor-specific molecular targets for inhibition of cell proliferation.


Assuntos
Citocinese/genética , Microtúbulos/genética , Septinas/genética , Estatmina/genética , Animais , Proliferação de Células/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Gástrula/crescimento & desenvolvimento , Humanos , Camundongos , Compostos de Fenilureia/farmacologia , Piridinas/farmacologia , Septinas/biossíntese , Deleção de Sequência , Estatmina/biossíntese
15.
Cell Signal ; 26(7): 1532-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24686088

RESUMO

Stathmin is an oncoprotein and is expressed at high levels in a wide variety of human malignancies, which plays important roles in maintenance of malignant phenotypes. The regulation of Stathmin gene overexpression has been wildly explored, but the exact mechanism still needs to be elucidated. It is believed that regulation of an oncogene protein abundance through post-translational modifications is essential for maintenance of malignant phenotypes. Here we identified the Rlim, a Ring H2 zinc finger protein with intrinsic ubiquitin ligase activity, as a Stathmin-interacting protein that could increase Stathmin turnover through binding with this targeted protein and then induce its degradation by proteasome in a ubiquitin-dependent manner. Inhibition of endogenous Rlim expression by siRNA could increase the level of Stathmin protein, which further led to cell proliferation and cell cycle changes in human osteosarcoma cell lines. On the other hand, forced overexpression of Rlim could decrease the level of Stathmin protein. These results demonstrate that Rlim is involved in the negative regulation of Stathmin protein level through physical interaction and ubiquitin-mediated proteolysis. Hence, Rlim is a novel regulator of Stathmin protein in a ubiquitin-dependent manner, and represents a new pathway for malignant phenotype turnover by modulating the level of Stathmin protein in human osteosarcomas.


Assuntos
Neoplasias Ósseas/metabolismo , Osteossarcoma/metabolismo , Estatmina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Células HEK293 , Humanos , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteólise , Interferência de RNA , RNA Interferente Pequeno , Estatmina/biossíntese , Ubiquitina/química , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
16.
Tumour Biol ; 35(3): 2619-29, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24218338

RESUMO

Data mining on public domain identified that stathmin 1 (STMN1) transcript was significantly higher expressed in nasopharyngeal carcinoma (NPC). Also known as the oncoprotein 18, STMN1 performs an important function in regulating rapid microtubule remodeling of the cytoskeleton in response to the cellular conditions. Immunoexpression of STMN1 was retrospectively assessed in biopsies of 124 consecutive NPC patients without initial distant metastasis and treated with consistent guidelines. The outcome was correlated with clinicopathological features and patient survivals. Results indicated that high STMN1 expressions (50 %) were correlated with advanced age (p = 0.027), higher T stage (p = 0.003), and overall clinical stage (p = 0.006) by the 7th American Joint Committee of Cancer Staging. In multivariate analyses, high STMN1 expression emerged as an independent prognosticator for worse disease-specific survival (p = 0.001), distal metastasis-free survival (p = 0.003), and local recurrence-free survival (p = 0.006). Exogenous expression of E2F transcription factor 1 (E2F1) or/and its dimeric partner, transcription factor Dp-1 (TFDP1), notably induced the STMN1 protein level in a NPC-derived cell line, TW01. Accordingly, high STMN1 protein level is commonly associated with adverse prognosticators and confers tumor aggressiveness in patients with NPC, and its upregulation might be attributed to E2F1 and/or TFDP1 transactivation.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/mortalidade , Estatmina/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma , Intervalo Livre de Doença , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patologia , Estadiamento de Neoplasias , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatmina/análise , Transfecção , Adulto Jovem
17.
J Immunol ; 191(12): 6250-60, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24227773

RESUMO

Recent research has indicated a new mode of intercellular communication facilitated by the movement of RNA between cells. There is evidence that RNA can transfer between cells in a multitude of ways, including in complex with proteins or lipids or in vesicles, including apoptotic bodies and exosomes. However, there remains little understanding of the function of nucleic acid transfer between human cells. In this article, we report that human macrophages transfer microRNAs (miRNAs) to hepato-carcinoma cells (HCCs) in a manner that required intercellular contact and involved gap junctions. Two specific miRNAs transferred efficiently between these cells--miR-142 and miR-223--and both were endogenously expressed in macrophages and not in HCCs. Transfer of these miRNAs influenced posttranscriptional regulation of proteins in HCCs, including decreased expression of reporter proteins and endogenously expressed stathmin-1 and insulin-like growth factor-1 receptor. Importantly, transfer of miRNAs from macrophages functionally inhibited proliferation of these cancerous cells. Thus, these data led us to propose that intercellular transfer of miRNA from immune cells could serve as a new defense against unwanted cell proliferation or tumor growth.


Assuntos
Carcinoma Hepatocelular/patologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/patologia , Macrófagos/metabolismo , MicroRNAs/fisiologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Comunicação Celular , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Linhagem Celular Tumoral , Micropartículas Derivadas de Células , Técnicas de Cocultura , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/fisiologia , Exossomos , Genes Reporter , Células Hep G2 , Humanos , Leucemia Monocítica Aguda/patologia , Mastocitoma/patologia , MicroRNAs/biossíntese , MicroRNAs/genética , RNA Interferente Pequeno/farmacologia , Receptor IGF Tipo 1/biossíntese , Receptor IGF Tipo 1/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Estatmina/biossíntese , Estatmina/genética , Tiazolidinas/farmacologia , Transfecção
18.
Cancer Biomark ; 13(1): 59-66, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23736022

RESUMO

Currently, the transcript abundance of key enzymes for chemotherapy drug metabolism, which may help in predicting the efficacy of a drug, can easily be detected in tumor tissues. However, there has been little research on the enzymes involved in the chemotherapy of gliomas. This study aimed to detect and compare the abundance of glioma chemotherapy drug-associated marker molecules in both gliomas and normal brain tissues and among gliomas of different grades. We examined the transcript abundance of four such marker molecules, MGMT, ERCC1, Topo IIα and Stathmin, in 46 glioma and 6 normal brain tissues. We also compared the abundance of these molecules in normal brain tissues and glioma tissues with different malignancy grades. Furthermore, we described the variation of these molecules in different grades of gliomas by calculating the ratio of their maximum to their minimum. The transcript abundance of MGMT and ERCC1 was significantly higher in normal brain tissues than in glioma tissues. However, the opposite result was observed for Topo IIα. For Stathmin, no significant differences between normal brain tissues and gliomas tissues were found. For all 4 marker molecules, no significant differences were detected between grades of glioma. All four molecules exhibited wide variation in abundance, fluctuating significantly between gliomas. These results suggest that individualized detection and medication may be beneficial for treatment.


Assuntos
Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Glioma/enzimologia , Glioma/genética , Adolescente , Adulto , Idoso , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Encéfalo/enzimologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Estudos de Casos e Controles , Criança , Metilases de Modificação do DNA/biossíntese , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/biossíntese , Enzimas Reparadoras do DNA/genética , DNA Topoisomerases Tipo II/biossíntese , DNA Topoisomerases Tipo II/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Endonucleases/biossíntese , Endonucleases/genética , Feminino , Glioma/tratamento farmacológico , Glioma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Estatmina/biossíntese , Estatmina/genética , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Adulto Jovem
19.
Oncogene ; 32(10): 1330-7, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22665054

RESUMO

In previous studies, we demonstrated that miR-193b expression is reduced in melanoma relative to benign nevi, and also that miR-193b represses cyclin D1 and Mcl-1 expression. We suggested that stathmin 1 (STMN1) might be a target of miR-193b. STMN1 normally regulates microtubule dynamics either by sequestering free tubulin heterodimers or by promoting microtubule catastrophe. Increased expression of STMN1 has been observed in a variety of human malignancies, but its association with melanoma is unknown. We now report that STMN1 is upregulated during the progression of melanoma relative to benign nevi, and that STMN1 is directly regulated by miR-193b. Using an experimental cell culture approach, overexpression of miR-193b using synthetic microRNAs repressed STMN1 expression, whereas inhibition of miR-193b with anti-miR oligos increased STMN1 expression in melanoma cells. The use of a luciferase reporter assay confirmed that miR-193b directly regulates STMN1 by targeting the 3'-untranslated region of STMN1 mRNA. We further demonstrated that STMN1 is overexpressed in malignant melanoma compared with nevi in two independent melanoma cohorts, and that its level is inversely correlated with miR-193b expression. However, STMN1 expression was not significantly associated with patient survival, Breslow depth, mitotic count or patient age. STMN1 knockdown by small-interfering RNA in melanoma cells drastically repressed cell proliferation and migration potential, whereas ectopic expression of STMN1 using lentivirus increased cell proliferation and migration rates. Subsequent gene expression analysis indicated that interconnected cytoskeletal networks are directly affected following STMN1 knockdown. In addition, we identified deregulated genes associated with proliferation and migration, and revealed that p21(Cip1/Waf1) and p27(Kip) could be downstream effectors of STMN1 signaling. Taken together, our study suggests that downregulation of miR-193b may contribute to increased STMN1 expression in melanoma, which consequently promotes migration and proliferation of tumor cells.


Assuntos
Melanoma/genética , Estatmina/genética , Regiões 3' não Traduzidas , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Oncogenes , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Estatmina/biossíntese , Estatmina/metabolismo , Transfecção , Regulação para Cima
20.
Biochim Biophys Acta ; 1819(11-12): 1164-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23000483

RESUMO

Stathmin (STMN1) is a microtubule destabilizing protein with a key role in cell cycle progression and cell migration that is up-regulated in several cancers and may contribute to the malignant phenotype. However, the factors that regulate its expression are not well understood. Loss as well as gain-of-function p53 mutations up-regulate STMN1 and in acute myelogenous leukemia where p53 is predominantly wild-type, STMN1 is also over-expressed. Here we show regulatory control of STMN1 expression by the leucine zipper transcription factor (TF) CREB1 and the basic helix-loop-helix TF LYL1. By ChIP-chip experiments we demonstrate in vivo the presence of LYL1 and CREB1 in close proximity on the STMN1 promoter and using promoter assays we reveal co-regulation of STMN1 by CREB1 and LYL1. By contrast, TAL1, another suspected oncoprotein in leukemia and close relative of LYL1, exerts no regulatory effect on the STMN1 promoter. NLI, LMO2 and GATA2 are previously described co-activators of Tal1/Lyl1-E47 transcriptional complexes and potentiate Lyl1 activation of the STMN1 promoter while having no effect on TAL1 transactivation. Promoter mutations that abrogate CREB1 proximal binding or mutations of the DNA-binding domain of CREB1 abolish LYL1 transcriptional activation. These results show that CRE and Ebox sites function as coordinated units and support previous evidence of joint CREB1-and LYL1 transcription events activating an aberrant subset of promoters in leukemia. CREB1 or LYL1 shRNA knock-down down-regulate STMN1 expression. Because down-regulation of STMN1 has been shown to have anti-proliferative effects, while CREB1 and LYL1 are suspected oncoproteins, interference with CREB1-LYL1 interactions may complement standard chemotherapy and yield additional beneficial effects.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação Leucêmica da Expressão Gênica , Leucemia/metabolismo , Proteínas de Neoplasias/metabolismo , Elementos de Resposta , Estatmina/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Técnicas de Silenciamento de Genes , Humanos , Células K562 , Leucemia/genética , Leucemia/patologia , Proteínas de Neoplasias/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Estatmina/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...