Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Plasmid ; 98: 37-44, 2018 06.
Article in English | MEDLINE | ID: mdl-30196057

ABSTRACT

The CRISPR/Cas9 system is a powerful genome editing tool for disrupting the expression of specific genes in a variety of cells. However, the genome editing procedure using currently available vectors is laborious, and there is room for improvement to obtain knockout cells more efficiently. Therefore, we constructed a novel vector for high efficiency genome editing, named pGedit, which contains EGFP-Bsr as a selection marker, expression units of Cas9, and sgRNA without a terminator sequence of the U6 promoter. EGFP-Bsr is a fusion protein of EGFP and blasticidin S deaminase, and enables rapid selection and monitoring of transformants, as well as confirmation that the vector has not been integrated into the genome. By using pGedit, we targeted human ACTB, ACTG1 and mouse Nes genes coding for ß-actin, γ-actin and nestin, respectively. Knockout cell lines of each gene were easily and efficiently obtained in all three cases. In this report, we show that our novel vector, pGedit, significantly facilitates genome editing.


Subject(s)
Actins/antagonists & inhibitors , CRISPR-Cas Systems , Gene Editing/methods , Genetic Vectors , Nestin/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Actins/genetics , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Base Sequence , Gene Targeting , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Nestin/genetics , Promoter Regions, Genetic , Sequence Homology
2.
J Huazhong Univ Sci Technolog Med Sci ; 37(6): 895-903, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29270750

ABSTRACT

Human Nestin (hNestin) has been found to express in melanoma, and its expression is positively correlated with the advanced stage of melanoma. However, the precise role of hNestin in the development of melanoma has not been fully understood. The present study aimed to explore the role of hNestin in the proliferation and invasion of melanoma cells. The lentivirus vector carrying a short hairpin RNAs (shRNAs) targeting hNestin (hNestin-shRNA-LV) was stably infected into human melanoma cells UACC903, which expressed high levels of hNestin. The effects of hNestin knockdown on the proliferation, apoptosis, migration of melanoma cells and the related signaling pathways were investigated by immunofluorence, Western blotting and reverse transcription polymerase chain reaction (RT-PCR), respectively. The results showed that hNestin was expressed in most melanoma specimens and the melanoma cells studied. Knockdown of hNestin expression significantly inhibited the proliferation of melanoma cells, blocked the formation of cell colony, arrested cell cycle at G1/S stage and suppressed the activation of Akt and GSK3ß. hNestin-silent cells also showed a sheet-like appearance with tight cell-cell adhesion, decreased membrane expression of N-cadherin and ß-catenin, and attenuated migration. Furthermore, hNestin silence resulted in the inhibition of tumor growth in vivo. Our study indicates that hNestin knockdown suppresses the proliferation of melanoma cells, which might be through affecting Akt-GSK3ß-Rb pathway-mediated G1/S arrest, and hNestin silence inhibits the migration by selectively modulating the expression of cell adhesion molecules in the process of epithelial-mesenchymal transition.


Subject(s)
Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/genetics , Melanoma/genetics , Nestin/genetics , Proto-Oncogene Proteins c-akt/genetics , Retinoblastoma Protein/genetics , Skin Neoplasms/genetics , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , G1 Phase Cell Cycle Checkpoints , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Melanoma/metabolism , Melanoma/pathology , Melanoma/therapy , Nestin/antagonists & inhibitors , Nestin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Tumor Burden , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
3.
Biochem Pharmacol ; 138: 1-6, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28389227

ABSTRACT

Sonic hedgehog (Shh) signaling plays a key role in regulation of normal development. The negative feedback mechanism mediated by the transcriptional factor, Gli3, acts to finely tune Shh signaling, providing tight control of normal developmental processes. Hyperactivation of Shh signaling often leads to many human malignancies, including basal cell carcinoma and medulloblastoma (MB). However, how tumor cells sustain the aberrant activation of Shh signaling is still not completely understood. We recently revealed that during MB formation, tumor cells express Nestin, a type VI intermediate filament protein, which maintains uncontrolled Shh signaling by abolishing negative feedback by Gli3. Therefore, Nestin expression is a necessary step for MB formation. These findings highlight the novel function of Nestin in regulating Shh signaling, as well as the important role of a disrupted negative feedback mechanism in MB tumorigenesis. Further, restoration of the intrinsic negative feedback by repressing Nestin expression represents a promising approach to treat MB as well as other Shh signaling associated malignancies.


Subject(s)
Antineoplastic Agents/therapeutic use , Cerebellar Neoplasms/drug therapy , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Medulloblastoma/drug therapy , Models, Biological , Nerve Tissue Proteins/metabolism , Nestin/antagonists & inhibitors , Animals , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/physiopathology , Disease Progression , Feedback, Physiological/drug effects , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Medulloblastoma/physiopathology , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nestin/metabolism , Signal Transduction/drug effects , Tumor Burden , Zinc Finger Protein Gli3
4.
Oncotarget ; 7(52): 87052-87063, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27894083

ABSTRACT

The expression of intermediate filament Nestin is necessary for the neural progenitor cells (NPCs) to maintain stemness, but the underlying cellular and molecular mechanism remains unclear. In this study, we demonstrated that Nestin is required for the self-renew of NPCs through activating MAPK and EGFR pathways. Knockdown of Nestin by shRNA inhibited cell cycle progression and proliferation in mouse NPCs. Moreover, suppression of Nestin reduced expression of the epidermal growth factor receptor (EGFR) in NPCs and inhibited the mitogenic effects of EGF on these cells. Treatment of NPCs with p38-MAPK inhibitor PD169316 reversed cell cycle arrest caused by the knockdown of Nestin. Our findings indicate that Nestin promotes NPC proliferation via p38-MAPK and EGFR pathways, and reveals the necessity of these pathways in NPCs self-renewal.


Subject(s)
ErbB Receptors/physiology , Nestin/physiology , Neural Stem Cells/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cell Cycle Checkpoints , Cell Differentiation , Cell Proliferation , Imidazoles/pharmacology , Mice , Nestin/antagonists & inhibitors , Neural Stem Cells/cytology , RNA, Small Interfering/genetics , Signal Transduction/physiology
5.
Oncogene ; 35(24): 3139-50, 2016 06 16.
Article in English | MEDLINE | ID: mdl-26434586

ABSTRACT

Nestin is widely expressed in numerous tumors and has become a diagnostic and prognostic indicator. However, the exact mechanism by which nestin contributes to tumor malignancy remains poorly understood. Here, we found marked upregulation of nestin expression in highly proliferative and invasive gastrointestinal stromal tumor (GIST) specimens. Nestin knockdown in GIST cells reduced the proliferative and invasive activity owing to a decrease of mitochondrial intracellular reactive oxygen species (ROS) generation. Furthermore, nestin was co-localized with mitochondria, and knockdown of nestin increased mitochondrial elongation and influenced the mitochondrial function, including oxygen consumption rates, ATP generation and mitochondrial membrane potential and so on. In exploring the underlying mechanism, we demonstrated nestin knockdown inhibited the mitochondrial recruitment of Dynamin-related protein1 and induced the change of mitochondrial dynamics. Thus, nestin may have an important role in GIST malignancy by regulating mitochondrial dynamics and altering intracellular ROS levels. The findings provide new clues to reveal mechanisms by which nestin mediates the proliferation and invasion of GISTs.


Subject(s)
Gastrointestinal Stromal Tumors/metabolism , Mitochondrial Dynamics/physiology , Nestin/metabolism , Animals , Cell Proliferation/physiology , Down-Regulation , Dynamins , Female , GTP Phosphohydrolases/metabolism , Gastrointestinal Stromal Tumors/pathology , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/metabolism , Middle Aged , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Invasiveness , Nestin/antagonists & inhibitors , Nestin/deficiency , Nestin/genetics , Prognosis , Reactive Oxygen Species/metabolism , Transfection
6.
Asian Pac J Cancer Prev ; 16(7): 2777-83, 2015.
Article in English | MEDLINE | ID: mdl-25854362

ABSTRACT

BACKGROUND: Nestin is associated with neoplastic transformation. However, the mechanisms by which nestin contributes regarding invasion and malignancy of gastric adenocarcinoma (GAC) remain unknown. Recent studies have shown that the epithelial-mesenchymal transition (EMT) is important in invasion and migration of cancer cells. In the present study, we aimed to investigate the expression of nestin and its correlation with EMT-related proteins in GAC. MATERIALS AND METHODS: The expression of nestin and EMT-related proteins was examined in GAC specimens and cell lines by immunohistochemistry and Western blotting. Clinicopathological features and survival outcomes were retrospectively analyzed. RESULTS: Positive nestin immunostaining was most obviously detected in the cytoplasm, nucleus or both cytoplasm and nucleus of tumor cells in 19.2% (24/125) of GAC tissues, which was significantly higher than that in normal gastric mucosa tissues (1.7%, 1/60) (p=0.001). Nestin expression was closely related to several clinicopathological factors and EMT-related proteins (E-cadherin, vimentin and Snail) and displayed a poor prognosis. Interestingly, simultaneous cytoplasmic and nuclear nestin expression correlated with EMT-related proteins (E-cadherin, vimentin and Snail) (p<0.05) and lymph node metastasis (p=0.041) and a shorter survival time (p<0.05), but this was not the case with cytoplasmic or nuclear nestin expression. CONCLUSIONS: Nestin, particularly expression in both cytoplasm and nucleus, might be involved in regulating EMT and malignant progression in GAC, with potential as an unfavorable indicator in tumor diagnosis and a target for clinical therapy.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Epithelial-Mesenchymal Transition , Nestin/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Staging , Nestin/antagonists & inhibitors , Nestin/genetics , Prognosis , RNA, Small Interfering/genetics , Retrospective Studies , Stomach Neoplasms/mortality , Survival Rate
7.
Cell Death Dis ; 5: e1377, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25118937

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a major malignant tumor of the head and neck region in southern China. The understanding of its underlying etiology is essential for the development of novel effective therapies. We report for the first time that microRNA-940 (miR-940) significantly suppresses the proliferation of a variety of cancer cell lines, arrests cells cycle, induces caspase-3/7-dependent apoptosis and inhibits the formation of NPC xenograft tumors in mice. We further show that miR-940 directly binds to the 3'-untranslated regions of Nestin mRNA and promotes its degradation. Likewise, depletion of Nestin inhibits tumor cell proliferation, arrest cells at G2/M, induces apoptosis and suppresses xenograft tumor formation in vivo. These functions of miR-940 can be reversed by ectopic expression of Nestin, suggesting that miR-940 regulates cell proliferation and survival through Nestin. Notably, we observed reduced miR-940 and increased Nestin levels in NPC patient samples. Protein microarray revealed that knockdown of Nestin in 5-8F NPC cells alters the phosphorylation of proteins involved in the DNA damage response, suggesting a mechanism for the miR-940/Nestin axis. Consistently, depletion of Nestin induced spontaneous DNA damage accumulation, delayed the DNA damage repair process and increased the sensitivity to irradiation and the chemotherapeutic agent doxorubicin. Collectively, our findings indicate that Nestin, which is downregulated by miR-940, can promote tumorigenesis in NPC cells through involvement in the DNA damage response. The levels of microRNA-940 and Nestin may serve as indicators of cancer status and prognosis.


Subject(s)
DNA Damage , MicroRNAs/metabolism , Nasopharyngeal Neoplasms/pathology , Nestin/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Base Sequence , Carcinoma , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gamma Rays , Humans , M Phase Cell Cycle Checkpoints/drug effects , Mice , Mice, Nude , MicroRNAs/genetics , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/metabolism , Nestin/antagonists & inhibitors , Nestin/genetics , Sequence Alignment , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...