Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Med ; 12(14): 15317-15336, 2023 07.
Article in English | MEDLINE | ID: mdl-37326412

ABSTRACT

PURPOSE: Flap endonuclease 1 (FEN1) is highly upregulated in prostate cancer and promotes the growth of prostate cancer cells. Androgen receptor (AR) is the most critical determinant of the occurrence, progression, metastasis, and treatment of prostate cancer. However, the effect of FEN1 on docetaxel (DTX) sensitivity and the regulatory mechanisms of AR on FEN1 expression in prostate cancer need to be further studied. METHODS: Bioinformatics analyses were performed using data from the Cancer Genome Atlas and the Gene Expression Omnibus. Prostate cancer cell lines 22Rv1 and LNCaP were used. FEN1 siRNA, FEN1 overexpression plasmid, and AR siRNA were transfected into cells. Biomarker expression was evaluated by immunohistochemistry and Western blotting. Apoptosis and the cell cycle were explored using flow cytometry analysis. Luciferase reporter assay was performed to verify the target relationship. Xenograft assays were conducted using 22Rv1 cells to evaluate the in vivo conclusions. RESULTS: Overexpression of FEN1 inhibited cell apoptosis and cell cycle arrest in the S phase induced by DTX. AR knockdown enhanced DTX-induced cell apoptosis and cell cycle arrest at the S phase in prostate cancer cells, which was attenuated by FEN1 overexpression. In vivo experiments showed that overexpression of FEN1 significantly increased tumour growth and weakened the inhibitory effect of DTX on prostate tumour growth, while AR knockdown enhance the sensitivity of DTX to prostate tumour. AR knockdown resulted in FEN1, pho-ERK1/2, and pho-ELK1 downregulation, and the luciferase reporter assay confirmed that ELK1 can regulate the transcription of FEN1. CONCLUSION: Collectively, our studies demonstrate that AR knockdown improves the DTX sensitivity of prostate cancer cells by downregulating FEN1 through the ERK/ELK1 signalling pathway.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Male , Humans , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , MAP Kinase Signaling System , Flap Endonucleases/genetics , Flap Endonucleases/metabolism , Cell Proliferation , Cell Line, Tumor , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Docetaxel/pharmacology , RNA, Small Interfering/metabolism , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism
2.
Adv Healthc Mater ; 11(23): e2201630, 2022 12.
Article in English | MEDLINE | ID: mdl-36148580

ABSTRACT

Neural stem cells (NSCs) transplantation has been considered as a promising strategy for the treatment of ischemic stroke. However, the therapeutic prospect is limited by the poor control over the survival, migration, and maturation of transplanted NSCs. Upregulating hypoxia inducible factor (HIF)-1α expression in stem cells can improve the survival and migration of NSCs grafted for stroke therapy. Functional peptide drugs, which could inhibit endogenous HIF-1α ubiquitination, might be used to effectively upregulate the HIF-1α expression in NSCs, thereby to improve the therapeutic effect in ischemia stroke. Herein, a magnetic resonance imaging (MRI)-visible nanomedicine is developed to codeliver functional peptides and superparamagnetic iron oxide (SPIO) nanoparticles into NSCs. This nanomedicine not only promotes the survival and migration ability of NSCs but also allows an in vivo tracking of transplanted NSCs with MRI. The results demonstrate the great potential of the functional peptides-complexed multifunctional nanomedicine in boosting the therapeutic effect of stem cell-based therapy in stroke.


Subject(s)
Neural Stem Cells , Stroke , Humans , Nanomedicine , Peptides , Magnetic Resonance Imaging , Stroke/diagnostic imaging , Stroke/drug therapy
3.
Nano Lett ; 21(1): 806-815, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33395306

ABSTRACT

Transplantation of neural stem cells (NSCs) is a promising treatment paradigm to replace lost neurons and reconstruct the damaged neural circuit after ischemic stroke. However, most transplanted NSCs often differentiate into astrocytes rather than functional neurons, and the poor neuronal differentiation adversely affects the therapeutic outcome of NSCs and limits their clinical translation for stroke therapy. Herein, a theranostic nanomedicine is developed to codeliver superparamagnetic iron oxide nanoparticles (SPIO) and small interfering RNA/antisense oligonucleotides (siRNA/ASO) against Pnky long noncoding RNA (lncRNA) into NSCs. This nanomedicine not only directs neuronal differentiation of NSCs through silencing the Pnky lncRNA but also allows an in vivo tracking of NSCs with magnetic resonance imaging. The enhanced neuronal differentiation of NSCs significantly improved the structural and functional recovery of the damaged brain after a stroke. The results demonstrate the great potential of the multifunctional nanomedicine targeting lncRNA to enhance stem cell-based therapies for a stroke.


Subject(s)
Neural Stem Cells , RNA, Long Noncoding , Stroke , Cell Differentiation , Humans , Nanomedicine , RNA, Long Noncoding/genetics , Stroke/diagnostic imaging , Stroke/genetics , Stroke/therapy
4.
Front Neurosci ; 14: 541, 2020.
Article in English | MEDLINE | ID: mdl-32536854

ABSTRACT

OBJECTIVE: Tinnitus is a prevalent hearing disorder, which could have a devastating impact on a patient's life. Functional studies have revealed connectivity pattern changes in the tinnitus brains that suggested a change of network dynamics as well as topological organization. However, no studies have yet provided evidence for the topological network changes in the gray matter. In this research, we aim to use the graph-theoretical approach to investigate the changes of topology in the tinnitus brain using structural MRI data, which could provide insights into the underlying anatomical basis for the neural mechanism in generating phantom sounds. METHODS: We collected 3D MRI images on 46 bilateral tinnitus patients and 46 age and gender-matched healthy controls. Brain networks were constructed with correlation matrices of the cortical thickness and subcortical volumes of 80 cortical/subcortical regions of interests. Global network properties were analyzed using local and global efficiency, clustering coefficient, and small-world coefficient, and regional network properties were evaluated using the betweenness coefficient for hub connectivity, and interregional correlations for edge properties. Between-group differences in cortical thickness and subcortical volumes were assessed using independent sample t-tests, and local efficiency, global efficiency, clustering coefficient, sigma, and interregional correlation were compared using non-parametric permutation tests. RESULTS: Tinnitus was found to have increased global efficiency, local efficiency, and cluster coefficient, indicating generally heightened connectivity of the network. The small-world coefficient remained normal for tinnitus, indicating intact small-worldness. Betweenness centrality analysis showed that hubs in the amygdala and parahippocampus were only found for tinnitus but not controls. In contrast, hubs in the auditory cortex, insula, and thalamus were only found for controls but not tinnitus. Interregional correlation analysis further found in tinnitus enhanced connectivity between the auditory cortex and prefrontal lobe, and decreased connectivity of the insula with anterior cingulate gyrus and parahippocampus. CONCLUSION: These findings provided the first morphological evidence of altered topological organization of the brain networks in tinnitus. These alterations suggest that heightened efficiency of the brain network and altered auditory-limbic connection for tinnitus, which could be developed in compensation for the auditory deafferentation, leading to overcompensation and, ultimately, an emotional and cognitive burden.

5.
Int J Cancer ; 142(5): 1033-1046, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29047121

ABSTRACT

Mesenchymal stem cells (MSCs) have emerged as a promising cellular vehicle for gene therapy of malignant gliomas due to their property of tumor tropism. However, MSCs may show bidirectional and divergent effects on tumor growth. Therefore, a robust surveillance system with a capacity for noninvasive monitoring of the homing, distribution and fate of stem cells in vivo is highly desired for developing stem cell-based gene therapies for tumors. In this study, we used ferritin gene-based magnetic resonance imaging (MRI) to track the tumor tropism of MSCs in a rat orthotopic xenograft model of malignant glioma. MSCs were transduced with lentiviral vectors expressing ferritin heavy chain (FTH) and enhanced green fluorescent protein (eGFP). Intra-arterial, intravenous and intertumoral injections of these FTH transgenic MSCs (FTH-MSCs) were performed in rats bearing intracranial orthotopic C6 gliomas. The FTH-MSCs were detected as hypointense signals on T2- and T2*-weighted images on a 3.0 T clinical MRI. After intra-arterial injection, 17% of FTH-MSCs migrated toward the tumor and gradually diffused throughout the orthotopic glioma. This dynamic process could be tracked in vivo by MRI up to 10 days of follow-up, as confirmed by histology. Moreover, the tumor tropism of MSCs showed no appreciable impact on the progression of the tumor. These results suggest that FTH reporter gene-based MRI can be used to reliably track the tropism and fate of MSCs after their systemic transplantation in orthotopic gliomas. This real-time in vivo tracking system will facilitate the future development of stem cell-based therapies for malignant gliomas.


Subject(s)
Brain Neoplasms/pathology , Ferritins/metabolism , Genes, Reporter , Glioma/pathology , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/pathology , Animals , Apoptosis , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Cell Proliferation , Glioma/metabolism , Glioma/therapy , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mesenchymal Stem Cells/metabolism , Rats , Rats, Wistar , Tropism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Int J Nanomedicine ; 12: 6705-6719, 2017.
Article in English | MEDLINE | ID: mdl-28932115

ABSTRACT

Understanding the long-term fate and potential mechanisms of mesenchymal stem cells (MSCs) after transplantation is essential for improving functional benefits of stem cell-based stroke treatment. Magnetic resonance imaging (MRI) is considered an attractive and clinically translatable tool for longitudinal tracking of stem cells, but certain controversies have arisen in this regard. In this study, we used SPION-loaded cationic polymersomes to label green fluorescent protein (GFP)-expressing MSCs to determine whether MRI can accurately reflect survival, long-term fate, and potential mechanisms of MSCs in ischemic stroke therapy. Our results showed that MSCs could improve the functional outcome and reduce the infarct volume of stroke in the brain. In vivo MRI can verify the biodistribution and migration of grafted cells when pre-labeled with SPION-loaded polymersome. The dynamic change of low signal volume on MRI can reflect the tendency of cell survival and apoptosis, but may overestimate long-term survival owing to the presence of iron-laden macrophages around cell graft. Only a small fraction of grafted cells survived up to 8 weeks after transplantation. A minority of these surviving cells were differentiated into astrocytes, but not into neurons. MSCs might exert their therapeutic effect via secreting paracrine factors rather than directing cell replacement through differentiation into neuronal and/or glial phenotypes.


Subject(s)
Brain Ischemia/diagnostic imaging , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Animals , Brain/cytology , Brain Ischemia/pathology , Brain Ischemia/therapy , Cell Differentiation , Cell Survival , Green Fluorescent Proteins/chemistry , Iron/metabolism , Male , Mesenchymal Stem Cells/chemistry , Neurons/cytology , Rats, Sprague-Dawley
7.
Nanomaterials (Basel) ; 7(5)2017 May 10.
Article in English | MEDLINE | ID: mdl-28489049

ABSTRACT

Cell-based therapy with mesenchymal stem cells (MSCs) is a promising strategy for acute ischemic stroke. In vivo tracking of therapeutic stem cells with magnetic resonance imaging (MRI) is imperative for better understanding cellular survival and migrational dynamics over time. In this study, we develop a novel biocompatible nanocomplex (ASP-SPIONs) based on cationic amylose, by introducing spermine and the image label, ultrasmall superparamagnetic iron oxide nanoparticles (SPIONs), to label MSCs. The capacity, efficiency, and cytotoxicity of the nanocomplex in transferring SPIONs into green fluorescence protein-modified MSCs were tested; and the performance of in vivo MRI tracking of the transplanted cells in acute ischemic stroke was determined. The results demonstrated that the new class of SPIONs-complexed nanoparticles based on biodegradable amylose can serve as a highly effective and safe carrier to transfer magnetic label into stem cells. A reliable tracking of transplanted stem cells in stroke was achieved by MRI up to 6 weeks, with the desirable therapeutic benefit of stem cells on stroke retained. With the advantages of a relatively low SPIONs concentration and a short labeling period, the biocompatible complex of cationic amylose with SPIONs is highly translatable for clinical application. It holds great promise in efficient, rapid, and safe labeling of stem cells for subsequent cellular MRI tracking in regenerative medicine.

8.
J Biomed Nanotechnol ; 12(12): 2112-24, 2016 Dec.
Article in English | MEDLINE | ID: mdl-29368886

ABSTRACT

MR imaging (MRI) upon cell labeling is an attractive and clinically translatable tool for longitudinally monitoring the survival and migration of stem cells. The common intracellular delivery of superparamagnetic iron oxide nanoparticles (SPIONs) via poly-L-lysine (PLL) requires a high SPION concentration and a long incubation period for appropriate cell labeling, which may negatively affect the viability and function of stem cells. In this study, we determined the performance of a new class of cationic polymersomes in transferring SPIONs into green fluorescence protein-modified mesenchymal stem cells (MSCs) for cellular MRI in acute ischemic stroke, compared with PLL-coated SPIONs. The results demonstrated that the polymersomes had comparable labeling efficiency and biological safety as well as a marginal benefit on post-transplantation cell survival; the polymersomes had the advantages of a relatively low SPION concentration and a substantially shorter labeling period compared with PLL-coated SPIONs. After transplantation, MSCs labeled using both methods offered a similar therapeutic effect on stroke, and cellular MRI could track the in vivo distribution and migration behavior of biologically active MSCs; however, MRI overestimated the true size of the cell grafts. SPION-loaded cationic polymersomes can be used as an alternative for the efficient, rapid, and safe labeling of stem cells for cellular MRI.


Subject(s)
Cell Tracking/methods , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Polylysine , Stem Cell Transplantation , Stroke/therapy , Animals , Cations , Cell Survival/drug effects , Green Fluorescent Proteins/chemistry , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity , Male , Polylysine/chemistry , Polylysine/toxicity , Rats , Rats, Sprague-Dawley , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...