Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell Tissue Bank ; 2023 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-37368142

RESUMO

Cerebrospinal fluid-contacting neurons (CSF-cNs) act crucial role in chemosensory and mechanosensory function in spinal cord. Recently, CSF-cNs were found to be an immature neuron and may be involved in spinal cord injury recovery. But how to culture it and explore its function in vitro are not reported in previous research. Here, we first reported culture and identification of CSF-cNs in vitro. We first established a protocol for in vitro culture of CSF-cNs from the cervical spinal cord of mice within 24 h after birth. Polycystic kidney disease 2-like 1 (PKD2L1)+ cells were isolated by fluorescence-activated cell sorting and expressed the neuron marker ß-tubulin III and CSF-cNs marker GABA. Intriguingly, PKD2L1+ cells formed neurosphere and expressed neural stem cell markers Nestin, Sox2 and GFAP. Thus, our research provided culture and isolation of CSF-cNs and this facilitate the investigation the CSF-cNs function in vitro.

2.
Front Cell Neurosci ; 16: 992520, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36159391

RESUMO

The neural stem cells (NSCs) in the ventricular-subventricular zone of the adult mammalian spinal cord may be of great benefit for repairing spinal cord injuries. However, the sources of NSCs remain unclear. Previously, we have confirmed that cerebrospinal fluid-contacting neurons (CSF-cNs) have NSC potential in vitro. In this study, we verified the NSC properties of CSF-cNs in vivo. In mouse spinal cords, Pkd2l1+ CSF-cNs localized around the central canal express NSC markers. In vitro, Pkd2l1+ CSF-cNs form a neurosphere and express NSC markers. Activation and proliferation of CSF-cNs can be induced by injection of the neurotrophic factors basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) into the lateral ventricle. Spinal cord injury (SCI) also induces NSC activation and proliferation of CSF-cNs. Collectively, our results demonstrate that Pkd2l1+ CSF-cNs have NSC properties in vivo and may be involved in SCI recovery.

3.
Onco Targets Ther ; 12: 11207-11220, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31908487

RESUMO

BACKGROUND: Metal regulatory transcription factor 2 (MTF2) has been previously reported as a protein binding to the metal response element of the mouse metallothionein promoter, which is involved in chromosome inactivation and pluripotency. However, the function of MTF2 in tumor formation and progression has not yet been completely elucidated. METHODS: The expression of MTF2 and clinicopathological characteristics were evaluated by hepatocellular carcinoma (HCC) tissue microarray of 240 specimens. The role of MTF2 on HCC progression was determined using MTT, crystal violet, and transwell assays. Tumor growth was monitored in a xenograft model, and intrahepatic metastasis models were established. RESULTS: The expression of MTF2 was increased in HCC and strongly associated with the clinical characteristics and prognosis. Forced expression of MTF2 in HCC cells significantly promoted cell growth, migration, and invasion in vitro. In contrast, downregulation of MTF2 inhibited cell growth, migration, and invasion in vitro. Moreover, knock down of MTF2 suppressed tumorigenesis and intrahepatic metastasis of HCC cells in vivo. Mechanistically, MTF2 overexpression may promote growth and epithelial-mesenchymal transition processes of HCC cells by facilitating Snail transcription. CONCLUSION: MTF2 promotes the proliferation, migration, and invasion of HCC cells by regulating Snail transcription, providing a potential therapeutic candidate for patients with HCC.

4.
Oncol Lett ; 15(6): 8749-8755, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29805613

RESUMO

Notch4, a family member of the Notch signaling pathway, has important roles in cellular developmental pathways, including proliferation, differentiation and apoptosis. The present study aimed to investigate the association between Notch4 expression and clinical outcomes with immunohistochemistry. Notch4 was expressed in 55.6% of triple-negative breast cancer (TNBC), 45.8% of Her-2-overexpressing and 25.5% of luminal breast cancer cases, with significantly higher expression occurring in TNBC (P<0.05). Furthermore, Notch4 expression was inversely associated with estrogen receptor (ER) and/or progesterone receptor positivity, and positively associated with larger tumor size, more lymph node involvement, and more advanced tumor node metastasis stage (P<0.05). No significant association was identified regarding age, menopausal status, Her-2 status or distant metastasis. Univariate survival analysis revealed that patients with low Notch4-expressing tumors exhibited a lower relative risk of cancer recurrence compared with patients with high Notch4-expressing tumors. However, in the luminal cohort, high Notch4 expression conferred significantly lower 5-year overall survival (OS) rates compared with Notch4 low-expression groups (P=0.003) but not in TNBC and Her-2-overexpressing patients. In conclusion, Notch4 expression was significantly higher in patients with TNBC and Her-2-overexpressing breast cancer compared with luminal breast cancer patients. Notch4 expression is associated with aggressive clinicopathological and biological phenotypes, and may predict poor prognosis in luminal breast cancer patients.

5.
NPJ Breast Cancer ; 4: 20, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30109262

RESUMO

Basal-like breast cancer (BLBC) is an aggressive subtype with a strong tendency to metastasize. Due to the lack of effective chemotherapy, BLBC has a poor prognosis compared with luminal subtype breast cancer. MicroRNA-221 and -222 (miR-221/222) are overexpressed in BLBC and associate with metastasis as well as poor prognosis; however, the mechanisms by which miR-221/222 function as oncomiRs remain unknown. Here, we report that miR-221/222 expression is inversely correlated with Notch3 expression in breast cancer cell lines. Notch3 is known to be overexpressed in luminal breast cancer cells and inhibits epithelial to mesenchymal transition (EMT). We demonstrate that miR-221/222 target Notch3 by binding to its 3' untranslated region and suppressing protein translation. Ectopic expression of miR-221/222 significantly promotes EMT, whereas overexpression of Notch3 intracellular domain attenuates the oncogenic function of miR-221/222, suggesting that miR-221/222 exerts its oncogenic role by negatively regulating Notch3. Taken together, our results elucidated that miR-221/222 promote EMT via targeting Notch3 in breast cancer cell lines suggesting that miR-221/222 can serve as a potential therapeutic target in BLBC.

6.
Oncogenesis ; 7(8): 59, 2018 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-30100605

RESUMO

Notch3 and GATA binding protein 3 (GATA-3) have been, individually, shown to maintain luminal phenotype and inhibit epithelial-mesenchymal transition (EMT) in breast cancers. In the present study, we report that Notch3 expression positively correlates with that of GATA-3, and both are associated with estrogen receptor-α (ERα) expression in breast cancer cells. We demonstrate in vitro and in vivo that Notch3 suppressed EMT and breast cancer metastasis by activating GATA-3 transcription. Furthermore, Notch3 knockdown downregulated GATA-3 and promoted EMT; while overexpression of Notch3 intracellular domain upregulated GATA-3 and inhibited EMT, leading to a suppression of metastasis in vivo. Moreover, inhibition or overexpression of GATA-3 partially reversed EMT or mesenchymal-epithelial transition induced by Notch3 alterations. In breast cancer patients, high GATA-3 expression is associated with higher Notch3 expression and lower lymph node metastasis, especially for hormone receptor (HR) positive cancers. Herein, we demonstrate a novel mechanism whereby Notch3 inhibit EMT by transcriptionally upregulating GATA-3 expression, at least in part, leading to the suppression of cancer metastasis in breast cancers. Our findings expand our current knowledge on Notch3 and GATA-3's roles in breast cancer metastasis.

7.
Theranostics ; 7(16): 4041-4056, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29109797

RESUMO

The luminal A phenotype is the most common breast cancer subtype and is characterized by estrogen receptor α expression (ERα). Identification of the key regulator that governs the luminal phenotype of breast cancer will clarify the pathogenic mechanism and provide novel therapeutic strategies for this subtype of cancer. ERα signaling pathway sustains the epithelial phenotype and inhibits the epithelial-mesenchymal transition (EMT) of breast cancer. In this study, we demonstrate that Notch3 positively associates with ERα in both breast cancer cell lines and human breast cancer tissues. We found that overexpression of Notch3 intra-cellular domain, a Notch3 active form (N3ICD), in ERα negative breast cancer cells re-activated ERα, while knock-down of Notch3 reduced ERα transcript and proteins, with alteration of down-stream genes, suggesting its ability to regulate ERα. Mechanistically, our results show that Notch3 specifically binds to the CSL binding element of the ERα promoter and activates ERα expression. Moreover, Notch3 suppressed EMT, while suppression of Notch3 promoted EMT in cellular assay. Overexpressing N3ICD in triple-negative breast cancer suppressed tumorigenesis and metastasis in vivo. Conversely, depletion of Notch3 in luminal breast cancer promoted metastasis in vivo. Furthermore, Notch3 transcripts were significantly associated with prolonged relapse-free survival in breast cancer, in particular in ERα positive breast cancer patients. Our observations demonstrate that Notch3 governs the luminal phenotype via trans-activating ERα expression in breast cancer. These findings delineate the role of a Notch3/ERα axis in maintaining the luminal phenotype and inhibiting tumorigenesis and metastasis in breast cancer, providing a novel strategy to re-sensitize ERα negative or low-expressing breast cancers to hormone therapy.


Assuntos
Neoplasias da Mama/metabolismo , Receptor Notch3/metabolismo , Receptores de Estrogênio/metabolismo , Neoplasias da Mama/genética , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor Notch3/genética , Receptores de Estrogênio/genética
8.
Cell Cycle ; 15(3): 432-40, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26694515

RESUMO

Uncontrolled cell proliferation, genomic instability and cancer are closely related to the abnormal activation of the cell cycle. Therefore, blocking the cell cycle of cancer cells has become one of the key goals for treating malignancies. Unfortunately, the factors affecting cell cycle progression remain largely unknown. In this study, we have explored the effects of Notch3 on the cell cycle in breast cancer cell lines by 3 methods: overexpressing the intra-cellular domain of Notch3 (N3ICD), knocking-down Notch3 by RNA interference, and using X-ray radiation exposure. The results revealed that overexpression of Notch3 arrested the cell cycle at the G0/G1 phase, and inhibited the proliferation and colony-formation rate in the breast cancer cell line, MDA-MB-231. Furthermore, overexpressing N3ICD upregulated Cdh1 expression and resulted in p27(Kip) accumulation by accelerating Skp2 degradation. Conversely, silencing of Notch3 in the breast cancer cell line, MCF-7, caused a decrease in expression levels of Cdh1 and p27(Kip) at both the protein and mRNA levels, while the expression of Skp2 only increased at the protein level. Correspondingly, there was an increase in the percentage of cells in the G0/G1 phase and an elevated proliferative ability and colony-formation rate, which may be caused by alterations of the Cdh1/Skp2/p27 axis. These results were also supported by exposing MDA-MB-231 cells or MCF-7 treated with siN3 to X-irradiation at various doses. Overall, our data showed that overexpression of N3ICD upregulated the expression of Cdh1 and caused p27(Kip) accumulation by accelerating Skp2 degradation, which in turn led to cell cycle arrest at the G0/G1 phase, in the context of proliferating breast cancer cell lines. These findings help to illuminate the precision therapy targeted to cell cycle progression, required for cancer treatment.


Assuntos
Caderinas/metabolismo , Receptor Notch3/metabolismo , Antígenos CD , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular , Humanos , Células MCF-7 , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Radiação Ionizante , Reação em Cadeia da Polimerase em Tempo Real , Receptor Notch3/antagonistas & inibidores , Receptor Notch3/genética , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Regulação para Cima/efeitos da radiação
9.
Oncotarget ; 6(25): 21704-17, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26023734

RESUMO

In human breast cancer, estrogen receptor-α (ERα) suppresses epithelial-mesenchymal transition (EMT) and stemness, two crucial parameters for tumor metastasis; however, the underlying mechanism by which ERα regulates these two processes remains largely unknown. Bmi1, the polycomb group protein B lymphoma Mo-MLV insertion region 1 homolog, regulates EMT transition, maintains the self-renewal capacity of stem cells, and is frequently overexpressed in human cancers. In the present study, ERα upregulated the expression of the epithelial marker, E-cadherin, in breast cancer cells through the transcriptional down-regulation of Bmi1. Furthermore, ERα overexpression suppressed the migration, invasion, and EMT of breast cancer cells. Notably, overexpression of ERα significantly decreased the CD44high/CD24low cell population and inhibited the capacity for mammosphere formation in ERα-negative breast cancer cells. In addition, overexpression of Bmi1 attenuated the ERα-mediated suppression of EMT and cell stemness. Immunohistochemistry revealed an inverse association of ERα and Bmi1 expression in human breast cancer tissue. Taken together, our findings suggest that ERα inhibits EMT and stemness through the downregulation of Bmi1.


Assuntos
Neoplasias da Mama/metabolismo , Transição Epitelial-Mesenquimal , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica , Complexo Repressor Polycomb 1/metabolismo , Animais , Antígeno CD24/metabolismo , Caderinas/metabolismo , Movimento Celular , Estrogênios/metabolismo , Feminino , Humanos , Receptores de Hialuronatos/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Complexo Repressor Polycomb 1/antagonistas & inibidores , Regiões Promotoras Genéticas , Ligação Proteica , Transdução de Sinais , Células-Tronco/citologia , Cicatrização
11.
Dalton Trans ; 40(4): 919-26, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21152487

RESUMO

The bis(pyridyl)-substituted TTF derivative, 2,6(7)-bis(4-pyridyl)-1,4,5,8-tetrathiafulvalene (TTF(py)(2)), and an inorganic analogue, [Ni(4-pedt)(2)] (4-pedt = 1-(pyridin-4-yl)ethylene-1,2-dithiolate), were used as bridging ligands to construct two multinuclear complexes {Co(II)(2)(Tp(Ph2))(2)(OAc)(2)[TTF(py)(2)]} (1, Tp(Ph2) = hydridotri(3,5-diphenylpyrazol-1-yl)borate) and {Co(II)(2)(Tp(Ph2))(2)(OAc)(2)[Ni(4-pedt)(2)]} (2), and two 1D zigzag chain complexes, {[M(II)(tta)(2)][TTF(py)(2)]}(n) (M = Cu for 3, and Mn for 4; tta = thenoyltrifluoroacetonate). X-Ray structural studies indicate that complexes 1 and 2 are very similar as a result of the isolobal analogy between TTF(py)(2) and [Ni(4-pedt)(2)], whereas complexes 3 and 4 are isostructural. The absorption spectra, electrochemical and magnetic properties for these new complexes have been studied. The results show that the interactions between the paramagnetic ions are weak owing to the large separation of the bridging ligands of TTFs and the inorganic analogue.

12.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 17(1): 229-33, 2009 Feb.
Artigo em Zh | MEDLINE | ID: mdl-19236786

RESUMO

Matrix metalloproteinases (MMPs) have the ability to degrade extracellular matrix components. They abnormally express in a variety of solid tumors and hematologic malignancies, and play an important role in tumor invasion and metastasis through extracellular matrix degradation, which closely relates with the progression and prognosis of the malignant disease. This article reviews progress of study on the mechanism of MMP underlying the pathogenesis of hematologic malignancies, including structure of MMP, regulation mechanism of MMP and its relation with proliferation and differentiation of hematopoietic stem cells (HSC), angiogenesis and tumor immunology and so on.


Assuntos
Neoplasias Hematológicas , Metaloproteinases da Matriz , Neoplasias Hematológicas/patologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA