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.
Stem Cell Res Ther ; 13(1): 160, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35410459

ABSTRACT

BACKGROUND: Breast carcinoma-amplified sequence 2 (BCAS2) regulates ß-catenin gene splicing. The conditional knockout of BCAS2 expression in the forebrain (BCAS2 cKO) of mice confers impaired learning and memory along with decreased ß-catenin expression. Because ß-catenin reportedly regulates adult neurogenesis, we wondered whether BCAS2 could regulate adult neurogenesis via ß-catenin. METHODS: BCAS2-regulating neurogenesis was investigated by characterizing BCAS2 cKO mice. Also, lentivirus-shBCAS2 was intracranially injected into the hippocampus of wild-type mice to knock down BCAS2 expression. We evaluated the rescue effects of BCAS2 cKO by intracranial injection of adeno-associated virus encoding BCAS2 (AAV-DJ8-BCAS2) and AAV-ß-catenin gene therapy. RESULTS: To show that BCAS2-regulating adult neurogenesis via ß-catenin, first, BCAS2 cKO mice showed low SRY-box 2-positive (Sox2+) neural stem cell proliferation and doublecortin-positive (DCX+) immature neurons. Second, stereotaxic intracranial injection of lentivirus-shBCAS2 knocked down BCAS2 in the hippocampus of wild-type mice, and we confirmed the BCAS2 regulation of adult neurogenesis via ß-catenin. Third, AAV-DJ8-BCAS2 gene therapy in BCAS2 cKO mice reversed the low proliferation of Sox2+ neural stem cells and the decreased number of DCX+ immature neurons with increased ß-catenin expression. Moreover, AAV-ß-catenin gene therapy restored neuron stem cell proliferation and immature neuron differentiation, which further supports BCAS2-regulating adult neurogenesis via ß-catenin. In addition, cells targeted by AAV-DJ8 injection into the hippocampus included Sox2 and DCX immature neurons, interneurons, and astrocytes. BCAS2 may regulate adult neurogenesis by targeting Sox2+ and DCX+ immature neurons for autocrine effects and interneurons or astrocytes for paracrine effects. CONCLUSIONS: BCAS2 can regulate adult neurogenesis in mice via ß-catenin.


Subject(s)
Neural Stem Cells , beta Catenin , Animals , Hippocampus , Mice , Neoplasm Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/metabolism , beta Catenin/genetics , beta Catenin/metabolism
2.
Neuron ; 103(5): 836-852.e5, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31277925

ABSTRACT

Polarized, non-overlapping, regularly spaced, tiled organization of radial glial cells (RGCs) serves as a framework to generate and organize cortical neuronal columns, layers, and circuitry. Here, we show that mediator of cell motility 1 (Memo1) is a critical determinant of radial glial tiling during neocortical development. Memo1 deletion or knockdown leads to hyperbranching of RGC basal processes and disrupted RGC tiling, resulting in aberrant radial unit assembly and neuronal layering. Memo1 regulates microtubule (MT) stability necessary for RGC tiling. Memo1 deficiency leads to disrupted MT minus-end CAMSAP2 distribution, initiation of aberrant MT branching, and altered polarized trafficking of key basal domain proteins such as GPR56, and thus aberrant RGC tiling. These findings identify Memo1 as a mediator of RGC scaffold tiling, necessary to generate and organize neurons into functional ensembles in the developing cerebral cortex.


Subject(s)
Ependymoglial Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Neocortex/embryology , Neural Stem Cells/metabolism , Neurons/metabolism , Animals , Autistic Disorder/genetics , Cell Movement/genetics , Cell Polarity , Cerebellum/embryology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Ependymoglial Cells/cytology , Gene Knockdown Techniques , HEK293 Cells , Hippocampus/embryology , Humans , Mice , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neocortex/cytology , Neocortex/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Protein Transport , Receptors, G-Protein-Coupled/metabolism
3.
Biomacromolecules ; 17(12): 3883-3892, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27936723

ABSTRACT

A novel drug delivery strategy featured with enhanced uptake of nanoparticles (NPs) by targeted tumor cells and subsequent intratumoral cellular hitchhiking of chemotherapy to deep tumor regions was described. The NP delivery system was obtained from assembly of poly(lactic acid-co-glycolic acid)-grafted hyaluronic acid (HA-g-PLGA) together with an anticancer drug, SN38, in aqueous phase, followed by implementing the NP surface with a layer of methoxypoly(ethylene glycol)-b-poly(histamine methacrylamide) (mPEG-b-PHMA) via hydrophobic association to improve the colloidal stability both in vitro and in vivo. Upon arrival of these PEGylated NPs at the acidic tumor site through the EPR effect, mPEG-b-PHMA became detached from the NP surface by the charge transition of the PHMA blocks from neutral (hydrophobic) to positively charged (hydrophilic) state via acid-induced protonation of their imidazole groups in tumor microenvironment. The exposure of HA shell on the naked NP thus resulted in enhanced uptake of NPs by CD44-expressed tumor cells, including cancer cells and tumor-associated macrophages (TAMs). Along with the TAMs being further chemotactically recruited by hypoxia cells, the engulfed nanotherapeutics was thus transported into the avascular area in which the anticancer action of chemotherapy occurred by virtue of the drug release alongside PLGA degradation, similar to those arising in other tumor nonhypoxia regions.


Subject(s)
Camptothecin/analogs & derivatives , Drug Delivery Systems , Hyaluronic Acid/chemistry , Macrophages/drug effects , Nanoparticles/administration & dosage , Prostatic Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Cell Survival/drug effects , Irinotecan , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Polyesters/chemistry , Polyglycolic Acid/chemistry , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
4.
Sci Rep ; 6: 34927, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713508

ABSTRACT

Breast carcinoma amplified sequence 2 (BCAS2) is a core component of the hPrP19 complex that controls RNA splicing. Here, we performed an exon array assay and showed that ß-catenin is a target of BCAS2 splicing regulation. The regulation of dendrite growth and morphology by ß-catenin is well documented. Therefore, we generated conditional knockout (cKO) mice to eliminate the BCAS2 expression in the forebrain to investigate the role of BCAS2 in dendrite growth. BCAS2 cKO mice showed a microcephaly-like phenotype with a reduced volume in the dentate gyrus (DG) and low levels of learning and memory, as evaluated using Morris water maze analysis and passive avoidance, respectively. Golgi staining revealed shorter dendrites, less dendritic complexity and decreased spine density in the DG of BCAS2 cKO mice. Moreover, the cKO mice displayed a short dendrite length in newborn neurons labeled by DCX, a marker of immature neurons, and BrdU incorporation. To further examine the mechanism underlying BCAS2-mediated dendritic malformation, we overexpressed ß-catenin in BCAS2-depleted primary neurons and found that the dendritic growth was restored. In summary, BCAS2 is an upstream regulator of ß-catenin gene expression and plays a role in dendrite growth at least partly through ß-catenin.


Subject(s)
Dendrites/metabolism , Dendrites/pathology , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Prosencephalon/abnormalities , Prosencephalon/metabolism , beta Catenin/metabolism , Animals , Behavior, Animal/physiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Doublecortin Protein , Exons , Female , Gene Expression Regulation, Developmental , Humans , MCF-7 Cells , Mice , Mice, Knockout , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Neoplasm Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Oligonucleotide Array Sequence Analysis , Phenotype , RNA Splicing
5.
PLoS One ; 10(6): e0130706, 2015.
Article in English | MEDLINE | ID: mdl-26091239

ABSTRACT

Previously, we showed that BCAS2 is essential for Drosophila viability and functions in pre-mRNA splicing. In this study, we provide strong evidence that BCAS2 regulates the activity of Delta-Notch signaling via Delta pre-mRNA splicing. Depletion of dBCAS2 reduces Delta mRNA expression and leads to accumulation of Delta pre-mRNA, resulting in diminished transcriptions of Delta-Notch signaling target genes, such as cut and E(spl)m8. Furthermore, ectopic expression of human BCAS2 (hBCAS2) and Drosophila BCAS2 (dBCAS2) in a dBCAS2-deprived fly can rescue dBCAS2 depletion-induced wing damage to the normal phenotypes. These rescued phenotypes are correlated with the restoration of Delta pre-mRNA splicing, which affects Delta-Notch signaling activity. Additionally, overexpression of Delta can rescue the wing deformation by deprivation of dBCAS2; and the depletion of dBCAS2 can restore the aberrant eye associated with Delta-overexpressing retinas; providing supporting evidence for the regulation of Delta-Notch signaling by dBCAS2. Taken together, dBCAS2 participates in Delta pre-mRNA splicing that affects the regulation of Delta-Notch signaling in Drosophila wing development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Neoplasm Proteins/metabolism , RNA Precursors/metabolism , Receptors, Notch/metabolism , Animals , Drosophila/growth & development , Drosophila Proteins/genetics , Eye/metabolism , Humans , Neoplasm Proteins/genetics , Phenotype , Plasmids/genetics , Plasmids/metabolism , RNA Precursors/genetics , RNA Splicing , Receptors, Notch/genetics , Signal Transduction , Wings, Animal/growth & development , Wings, Animal/metabolism
6.
RNA ; 19(2): 208-18, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23249746

ABSTRACT

Here, we show that dBCAS2 (CG4980, human Breast Carcinoma Amplified Sequence 2 ortholog) is essential for the viability of Drosophila melanogaster. We find that ubiquitous or tissue-specific depletion of dBCAS2 leads to larval lethality, wing deformities, impaired splicing, and apoptosis. More importantly, overexpression of hBCAS2 rescues these defects. Furthermore, the C-terminal coiled-coil domain of hBCAS2 binds directly to CDC5L and recruits hPrp19/PLRG1 to form a core complex for splicing in mammalian cells and can partially restore wing damage induced by knocking down dBCAS2 in flies. In summary, Drosophila and human BCAS2 share a similar function in RNA splicing, which affects cell viability.


Subject(s)
Apoptosis/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Neoplasm Proteins/metabolism , RNA Splicing/genetics , Wings, Animal/abnormalities , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Knockdown Techniques , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Larva/growth & development , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity , Phenotype , Promoter Regions, Genetic , Protein Interaction Mapping , Protein Structure, Tertiary , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins , Wings, Animal/growth & development
7.
Int J Biochem Cell Biol ; 44(11): 1952-61, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22846800

ABSTRACT

Damaged DNA-binding protein 2 (DDB2), a protein that binds damaged DNA, is a DDB1 and CUL4-associated factor. This study is the first to demonstrate that DDB2 is a novel androgen receptor (AR)-interacting protein; and mediating contact with AR and CUL4A-DDB1 complex for AR ubiquitination/degradation. DNA damage induces both p53 and DDB2 gene expression those two can inhibit AR expression. The former reduces AR via transcription regulation but the latter via proteosome degradation. Thereby DDB2 can inhibit cell growth rate in AR-expressing cells (LNCaP) but not in AR-null cells (PC3). Hence DDB2 may be a potential regimen for prostate cancer treatment, especially in androgen-refractory patients harboring high amount of AR who cannot be cured by androgen ablation.


Subject(s)
DNA-Binding Proteins/metabolism , Proteolysis , Receptors, Androgen/metabolism , Ubiquitination , Cell Line , Cell Proliferation , Cullin Proteins/metabolism , DNA Damage , DNA-Binding Proteins/chemistry , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Protein Interaction Mapping , Protein Stability , Protein Structure, Tertiary , Receptors, Androgen/genetics , Tumor Suppressor Protein p53/metabolism
8.
Cancer Res ; 69(23): 8877-85, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19903847

ABSTRACT

Breast cancer amplified sequence 2 (BCAS2) was reported previously as a transcriptional coactivator of estrogen receptor. Here, we report that BCAS2 directly interacts with p53 to reduce p53 transcriptional activity by mildly but consistently decreasing p53 protein in the absence of DNA damage. However, in the presence of DNA damage, BCAS2 prominently reduces p53 protein and provides protection against chemotherapeutic agent such as doxorubicin. Deprivation of BCAS2 induces apoptosis in p53 wild-type cells but causes G(2)-M arrest in p53-null or p53 mutant cells. There are at least two apoptosis mechanisms induced by silencing BCAS2 in wild-type p53-containing cells. Firstly, it increases p53 retention in nucleus that triggers the expression of apoptosis-related genes. Secondly, it increases p53 transcriptional activity by raising p53 phosphorylation at Ser(46) and decreases p53 protein degradation by reducing p53 phosphorylation at Ser(315). We show for the first time that BCAS2, a small nuclear protein (26 kDa), is a novel negative regulator of p53 and hence a potential molecular target for cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , DNA Damage , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...