Filters
Results 1 - 10 of 4686
Results 1 - 10 of 4686.
Search took: 0.026 seconds
Sort by: date | relevance |
Han, Bing; Fan, Jincai; Liu, Liqiang; Tian, Jia; Gan, Cheng; Yang, Zengjie; Jiao, Hu; Zhang, Tiran; Liu, Zheng; Zhang, Hua, E-mail: 1012980819@qq.com2019
AbstractAbstract
No abstract available
Primary Subject
Source
Copyright (c) 2019 Springer-Verlag London Ltd., part of Springer Nature; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Lasers in Medical Science (Online); ISSN 1435-604X;
; v. 34(1); p. 1-10

Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
Uranishi, Kousuke; Akagi, Tadayuki; Koide, Hiroshi; Yokota, Takashi, E-mail: tadayuki@staff.kanazawa-u.ac.jp, E-mail: tyokota@med.kanazawa-u.ac.jp2016
AbstractAbstract
[en] Estrogen-related receptor beta (Esrrb) is expressed in embryonic stem (ES) cells and is involved in self-renewal ability and pluripotency. Previously, we found that Dax1 is associated with Esrrb and represses its transcriptional activity. Further, the disruption of the Dax1–Esrrb interaction increases the expression of the extra-embryonic endoderm marker Gata6 in ES cells. Here, we investigated the influences of Esrrb and Dax1 on Gata6 expression. Esrrb overexpression in ES cells induced endogenous Gata6 mRNA and Gata6 promoter activity. In addition, the Gata6 promoter was found to contain the Esrrb recognition motifs ERRE1 and ERRE2, and the latter was the responsive element of Esrrb. Associations between ERRE2 and Esrrb were then confirmed by biotin DNA pulldown and chromatin immunoprecipitation assays. Subsequently, we showed that Esrrb activity at the Gata6 promoter was repressed by Dax1, and although Dax1 did not bind to ERRE2, it was associated with Esrrb, which directly binds to ERRE2. In addition, the transcriptional activity of Esrrb was enhanced by nuclear receptor co-activator 3 (Ncoa3), which has recently been shown to be a binding partner of Esrrb. Finally, we showed that Dax1 was associated with Ncoa3 and repressed its transcriptional activity. Taken together, the present study indicates that the Gata6 promoter is activated by Esrrb in association with Ncoa3, and Dax1 inhibited activities of Esrrb and Ncoa3, resulting maintenance of the undifferentiated status of ES cells. - Highlights: • Esrrb induced Gata6 expression in ES cells. • Gata6 promoter activity was enhanced by Esrrb, which was repressed by Dax1. • Dax1 associated with the Gata6 promoter via Esrrb. • Dax1 associated with Ncoa3 and repressed its transcriptional activity.
Primary Subject
Source
S0006-291X(16)31460-7; Available from http://dx.doi.org/10.1016/j.bbrc.2016.09.011; Copyright (c) 2016 Elsevier Science B.V., Amsterdam, The Netherlands, All rights reserved.; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Biochemical and Biophysical Research Communications; ISSN 0006-291X;
; CODEN BBRCA9; v. 478(4); p. 1720-1725

Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
Sethi, Aisha; Sholl, Lynette M., E-mail: lmsholl@partners.org2011
AbstractAbstract
[en] Cancer stem cells are defined as a subpopulation of cells within a tumor that are capable of self-renewal and differentiation into the heterogeneous cell lineages that comprise the tumor. Many studies indicate that cancer stem cells may be responsible for treatment failure and relapse in cancer patients. The factors that regulate cancer stem cells are not well defined. MicroRNAs (miRNAs) are small non-coding RNAs that regulate translational repression and transcript degradation. miRNAs play a critical role in embryonic and inducible pluripotent stem cell regulation and emerging evidence supports their role in cancer stem cell evolution. To date, miRNAs have been shown to act either as tumor suppressor genes or oncogenes in driving critical gene expression pathways in cancer stem cells in a wide range of human malignancies, including hematopoietic and epithelial tumors and sarcomas. miRNAs involved in cancer stem cell regulation provide attractive, novel therapeutic targets for cancer treatment. This review attempts to summarize progress to date in defining the role of miRNAs in cancer stem cells
Primary Subject
Source
Available from http://dx.doi.org/10.3390/cancers3043957; Available from http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3763404; PMCID: PMC3763404; PMID: 24213119; PUBLISHER-ID: cancers-03-03957; OAI: oai:pubmedcentral.nih.gov:3763404; Copyright (c) 2011 by the authors; licensee MDPI, Basel, Switzerland.; This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution license (http://creativecommons.org/licenses/by/3.0/).; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Cancers (Basel); ISSN 2072-6694;
; v. 3(4); p. 3957-3971

Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
External URLExternal URL
AbstractAbstract
[en] Epithelial-to-mesenchymal transition (EMT) is a process in which epithelial cells lose their cell-cell contacts resulting in the formation of mesenchymal cells with migratory properties. Increasing evidence indicate EMT plays a key role in the invasion, metastasis and therapeutic resistance of cancer and maintenance of the phenotype of cancer stem cells (CSCs), which makes the prognosis of patients worse. The progression of cancer from epithelial tissue towards a malignant phenotype is driven by multiple factors that remodel the tissue architecture. This review summarizes and analyzes current studies of genetic and microenvironmental factors in inducing and maintaining cancer EMT and therapeutic implications. This will enable a better understanding of the contribution of EMT-associated factors to cancer progression and highlights that genetic factors and tumor microenvironment responsible for EMT could be used as attractive targets for therapeutic intervention.
Primary Subject
Source
S0014482718306062; Available from http://dx.doi.org/10.1016/j.yexcr.2018.07.046; Copyright (c) 2018 Elsevier Inc. All rights reserved.; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
AbstractAbstract
[en] Cancer stem cells (CSCs) are cells within a tumor that possess the capacity to self-renew and maintain tumor-initiating capacity through differentiation into the heterogeneous lineages of cancer cells that comprise the whole tumor. These tumor-initiating cells could provide a resource for cells that cause tumor recurrence after therapy. Although the cell origin of CSCs remains to be fully elucidated, mounting evidence has demonstrated that Epithelial-to-Mesenchymal Transition (EMT), induced by different factors, is associated with tumor aggressiveness and metastasis and these cells share molecular characteristics with CSCs, and thus are often called cancer stem-like cells or tumor-initiating cells. The acquisition of an EMT phenotype is a critical process for switching early stage carcinomas into invasive malignancies, which is often associated with the loss of epithelial differentiation and gain of mesenchymal phenotype. Recent studies have demonstrated that EMT plays a critical role not only in tumor metastasis but also in tumor recurrence and that it is tightly linked with the biology of cancer stem-like cells or cancer-initiating cells. Here we will succinctly summarize the state-of-our-knowledge regarding the molecular similarities between cancer stem-like cells or CSCs and EMT-phenotypic cells that are associated with tumor aggressiveness focusing on solid tumors
Primary Subject
Source
Available from http://dx.doi.org/10.3390/cancers30100716; Available from http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3106306; PMCID: PMC3106306; PMID: 21643534; PUBLISHER-ID: cancers-03-00716; OAI: oai:pubmedcentral.nih.gov:3106306; Copyright (c) 2011 by the authors; licensee MDPI, Basel, Switzerland.; This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution license (http://creativecommons.org/licenses/by/3.0/).; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Cancers (Basel); ISSN 2072-6694;
; v. 3(1); p. 716-729

Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
External URLExternal URL
Fang, Tong-Jing; Wang, Ding-Han; Wang, Chia-Yu; Poongodi, Raju; Liou, Nien-Hsien; Liu, Jiang-Chuan; Hsu, Ming-Lun; Hong, Po-Da; Yang, Shih-Fang; Liu, Meng-Lun, E-mail: sfyang2017@gmail.com, E-mail: doc20054@gmail.com2018
AbstractAbstract
[en] The original version of this article unfortunately contained a mistake. The country was incorrect in the authors affiliations. It should read as “ROC”. The corrected affiliations are given below.
Primary Subject
Source
Copyright (c) 2018 Springer Science+Business Media, LLC, part of Springer Nature; http://www.springer-ny.com; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
AbstractAbstract
[en] AURKA (aurora kinase A) has been confirmed as an oncogene in cancer development; however, its role and underlying mechanisms in the metastasis of hepatocellular carcinoma (HCC) remain unknown. In this study, We found that AURKA was up-regulated in HCC tissues and correlated with pathological stage and distant metastasis. Further found that AURKA was involved in the cancer metastases after radiation in HCC. While overexpression of AURKA induced epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) behaviors though PI3K/AKT pathway, silencing AURKA suppressed radiation-enhanced cell invasiveness of HCC. Taken together, our results suggested that AURKA contributed in metastasis of irradiated residul HCC though facilitating EMT and CSC properties, suggesting the potential clinical application of AURKA inhibitors in radiotherapy for patients with HCC. - Highlights: • First reported overexpression of AURKA in HCC and correlation with poor OS. • AURKA was involved in the cancer metastases after radiation in HCC. • Further found AURKA promoted EMT and CSC behaviors though PI3K/AKT pathway. • Silencing AURKA suppressed radiation-enhanced cell invasiveness of HCC. • AURKA may be potential therapeutic target of HCC.
Primary Subject
Source
S0006-291X(17)30541-7; Available from http://dx.doi.org/10.1016/j.bbrc.2017.03.075; Copyright (c) 2017 Elsevier Science B.V., Amsterdam, The Netherlands, All rights reserved.; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Biochemical and Biophysical Research Communications; ISSN 0006-291X;
; CODEN BBRCA9; v. 486(2); p. 514-520

Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
AbstractAbstract
[en] Neuroblastoma (NB) is the most common extracranial solid tumor in children. NB tumors and derived cell lines are phenotypically heterogeneous. Cell lines are classified by phenotype, each having distinct differentiation and tumorigenic properties. The neuroblastic phenotype is tumorigenic, has neuronal features and includes stem cells (I-cells) and neuronal cells (N-cells). The non-neuronal phenotype (S-cell) comprises cells that are non-tumorigenic with features of glial/smooth muscle precursor cells. This study identified miRNAs associated with each distinct cell phenotypes and investigated their role in regulating associated differentiation and tumorigenic properties. A miRNA microarray was performed on the three cell phenotypes and expression verified by qRT-PCR. miRNAs specific for certain cell phenotypes were modulated using miRNA inhibitors or stable transfection. Neuronal differentiation was induced by RA; non-neuronal differentiation by BrdU. Changes in tumorigenicity were assayed by soft agar colony forming ability. N-myc binding to miR-375 promoter was assayed by chromatin-immunoprecipitation. Unsupervised hierarchical clustering of miRNA microarray data segregated neuroblastic and non-neuronal cell lines and showed that specific miRNAs define each phenotype. qRT-PCR validation confirmed that increased levels of miR-21, miR-221 and miR-335 are associated with the non-neuronal phenotype, whereas increased levels of miR-124 and miR-375 are exclusive to neuroblastic cells. Downregulation of miR-335 in non-neuronal cells modulates expression levels of HAND1 and JAG1, known modulators of neuronal differentiation. Overexpression of miR-124 in stem cells induces terminal neuronal differentiation with reduced malignancy. Expression of miR-375 is exclusive for N-myc-expressing neuroblastic cells and is regulated by N-myc. Moreover, miR-375 downregulates expression of the neuronal-specific RNA binding protein HuD. Thus, miRNAs define distinct NB cell phenotypes. Increased levels of miR-21, miR-221 and miR-335 characterize the non-neuronal, non-malignant phenotype and miR-335 maintains the non-neuronal features possibly by blocking neuronal differentiation. miR-124 induces terminal neuronal differentiation with reduction in malignancy. Data suggest N-myc inhibits neuronal differentiation of neuroblastic cells possibly by upregulating miR-375 which, in turn, suppresses HuD. As tumor differentiation state is highly predictive of patient survival, the involvement of these miRNAs with NB differentiation and tumorigenic state could be exploited in the development of novel therapeutic strategies for this enigmatic childhood cancer
Primary Subject
Secondary Subject
Source
Available from http://dx.doi.org/10.1186/1471-2407-14-309; Available from http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4038381; PMCID: PMC4038381; PUBLISHER-ID: 1471-2407-14-309; PMID: 24885481; OAI: oai:pubmedcentral.nih.gov:4038381; Copyright (c) 2014 Samaraweera et al.; licensee BioMed Central Ltd.; This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0) (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
BMC cancer (Online); ISSN 1471-2407;
; v. 14; p. 309

Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
External URLExternal URL
AbstractAbstract
[en] Bone is a rigid yet dynamic organ, and this dynamism is mediated by the delicate balance between osteoclastic bone resorption and osteoblastic bone formation. However, excessive activation of osteoclasts is responsible for many bone diseases such as osteoporosis, Paget disease, and tumor bone metastasis. Agents that could inhibit osteoclast formation or function are regarded as promising alternatives to treat osteoclast-related diseases. Recently, traditional Chinese medicine has attracted attention because of its multiple activities in bone metabolism. Among them, gastrodin has been reported as an anti-osteoporosis agent that reduces reactive oxygen species. However, the direct action of gastrodin on osteoclast differentiation and bone resorption, and its underlying molecular mechanism, remain unknown. In this study, we investigated the effects of gastrodin on receptor activator NF-κB ligand (RANKL)-activated osteoclasts formation and bone resorption. Our results showed that gastrodin retarded RANKL-induced osteoclast differentiation efficiently by downregulating transcriptional and translational expression of nuclear factor of activated T cells cl (NFATc1), a major factor in RANKL-mediated osteoclastogenesis. Meanwhile, gastrodin prevented osteoclast maturation and migration by inhibiting the gene expression of dendrocyte expressed seven transmembrane protein (DC-STAMP), an osteoclastic-specific gene that controls cells fusion and movement. And gastrodin prevented RANKL-induced osteoclastic bone erosion in vitro. In addition, gastrodin also stimulated bone mesenchymal stem cell (BMSC) spreading and osseointegration in titanium plate. In summary, gastrodin could prevent osteoclasts formation and bone resorption via blockage of NFATc1 activity, and stimulate osseointegration in vitro. Gastrodin could be developed as a potent phytochemical candidate to treat osteolytic diseases. - Highlights: • Gastrodin suppresses osteoclasts formation and function in vitro. • Gastrodin impairs NFATc1 activation. • Gastrodin stimulates osseointegration in vitro. • Gastrodin may be used for treating osteoclast related diseases.
Primary Subject
Source
S0006-291X(17)30254-1; Available from http://dx.doi.org/10.1016/j.bbrc.2017.01.179; Copyright (c) 2017 Elsevier Science B.V., Amsterdam, The Netherlands, All rights reserved.; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Biochemical and Biophysical Research Communications; ISSN 0006-291X;
; CODEN BBRCA9; v. 484(4); p. 820-826

Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
Kanungo, Jyotshna, E-mail: jyotshnabala.kanungo@fda.hhs.gov2017
AbstractAbstract
[en] RNA silencing is used as a common method for investigating loss-of-function effects of genes of interest. In mammalian cells, RNA interference (RNAi) or RNA silencing can be achieved by transient siRNA (small or short interfering RNA) transfection or by stable shRNA (short hairpin RNA) systems. Various vectors are used for efficient delivery of shRNA. Lentiviral vectors offer an efficient delivery system for stable and long-term expression of the shRNA in mammalian cells. The widely used lentiviral pLKO.1 plasmid vector is very popular in RNAi studies. A large RNAi database, a TRC (the RNAi Consortium) library, was established based on the pLKO.1-TRC plasmid vector. This plasmid (also called pLKO.1-puro) has a puromycin-resistant gene for selection in mammalian cells along with designs for generating lentiviral particles as well for RNA silencing. While using the pLKO.1-puro TRC control shRNA plasmid for transfection in murine P19 embryonic stem (ES) cells, it was unexpectedly discovered that this plasmid vector induced robust endodermal differentiation. Since P19 ES cells are pluripotent and respond to external stimuli that have the potential to alter the phenotype and thus its stemness, other cell types used in RNA silencing studies do not display the obvious effect and therefore, may affect experiments in subtle ways that would go undetected. This study for the first time provides evidence that raises concern and warrants extreme caution while using the pLKO.1-puro control shRNA vector because of its unexpected non-specific effects on cellular integrity. - Highlights: • In P19 ES cells the pLKO.1-puro lentiviral control shRNA vector induced endodermal differentiation. • P19 ES cells harboring the pCDNA3 plasmid vector retained their stem-ness as opposed to those harboring the pLKO.1-puro vector. • P19 ES cells can serve as a sensor to determine vector safety. • Extreme caution is warranted while using the widely used pLKO.1-puro lentiviral vector for experimental and therapeutic designs.
Primary Subject
Source
S0006-291X(17)30532-6; Available from http://dx.doi.org/10.1016/j.bbrc.2017.03.066; Copyright (c) 2017 Elsevier Science B.V., Amsterdam, The Netherlands, All rights reserved.; Country of input: International Atomic Energy Agency (IAEA)
Record Type
Journal Article
Journal
Biochemical and Biophysical Research Communications; ISSN 0006-291X;
; CODEN BBRCA9; v. 486(2); p. 481-485

Country of publication
Reference NumberReference Number
INIS VolumeINIS Volume
INIS IssueINIS Issue
1 | 2 | 3 | Next |