Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Radiat Res ; 193(3): 286-300, 2020 03.
Article in English | MEDLINE | ID: mdl-32017668

ABSTRACT

New radiosensitizers are urgently needed for radiation therapy patients with localized hepatocellular carcinoma (HCC) that is refractory to radical surgery. We previously found that genistein, a major soy isoflavone, exerts radioprotective effects on L-02 normal liver cells at low concentrations. Here, we report that 5 µM genistein shows less harm to L-02 cells than HCC cells and that it significantly enhances the radiosensitivity of HCC cells by enhancing DNA damage, chromosomal aberrations and cell cycle arrest at G2/M phase and by exacerbating apoptosis. Mechanistically, genistein aggravates X-ray-induced decreases in the levels of phospho-Bad (Ser136) but enhances the levels of phospho-Chk2 (Thr68), phospho-ATM (Ser1981) and γ-H2AX. Micro-array analysis indicated that downregulation of POU6F and CCNE2 expression and upregulation of FBXO32 and cyclin B1 expression might play vital roles in genistein-induced radiosensitivity. These findings suggest genistein as an interesting candidate for adjuvant radiotherapy for HCC and indicate that genistein causes less harm to normal cells than HCC cells by inducing G2/M arrest and apoptosis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Division/drug effects , G2 Phase/drug effects , Genistein/pharmacology , Liver Neoplasms/pathology , Radiation Tolerance/drug effects , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Humans , Mice , X-Rays
2.
Cell Res ; 29(3): 233-247, 2019 03.
Article in English | MEDLINE | ID: mdl-30728452

ABSTRACT

Mutations in RNA-binding proteins (RBPs) localized in ribonucleoprotein (RNP) granules, such as hnRNP A1 and TDP-43, promote aberrant protein aggregation, which is a pathological hallmark of various neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Protein posttranslational modifications (PTMs) are known to regulate RNP granules. In this study, we investigate the function of poly(ADP-ribosyl)ation (PARylation), an important PTM involved in DNA damage repair and cell death, in RNP granule-related neurodegeneration. We reveal that PARylation levels are a major regulator of the assembly-disassembly dynamics of RNP granules containing disease-related RBPs, hnRNP A1 and TDP-43. We find that hnRNP A1 can both be PARylated and bind to PARylated proteins or poly(ADP-ribose) (PAR). We further uncover that PARylation of hnRNP A1 at K298 controls its nucleocytoplasmic transport, whereas PAR-binding via the PAR-binding motif (PBM) of hnRNP A1 regulates its association with stress granules. Moreover, we reveal that PAR not only dramatically enhances the liquid-liquid phase separation of hnRNP A1, but also promotes the co-phase separation of hnRNP A1 and TDP-43 in vitro and their interaction in vivo. Finally, both genetic and pharmacological inhibition of PARP mitigates hnRNP A1- and TDP-43-mediated neurotoxicity in cell and Drosophila models of ALS. Together, our findings suggest a novel and crucial role for PARylation in regulating the dynamics of RNP granules, and that dysregulation in PARylation and PAR levels may contribute to ALS disease pathogenesis by promoting protein aggregation.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Poly ADP Ribosylation/physiology , Protein Aggregation, Pathological/genetics , Animals , Cell Line , DNA Damage/genetics , DNA Repair/genetics , Drosophila , Frontotemporal Dementia/pathology , HEK293 Cells , Humans , Mice , RNA-Binding Proteins/genetics
3.
Biochem Biophys Res Commun ; 505(2): 505-510, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30268497

ABSTRACT

Age-related cataract, the most common cause of blindness worldwide, has been found closely associated with ß-crystallin B2 (ßB2 or CRYBB2). MicroRNAs (miRNAs) are the primary epigenetic regulators important for various biological processes. However, the role of miRNAs in the progression of lens cataract remains to be elucidated. In this study, we found a novel signal cascade miR-326-fibroblast growth factor 1 (FGF1)-ßB2 modulating the progression of lens cataract. In brief, miR-326 exacerbated but its antagomirs attenuated H2O2-induced apoptosis of HLEC-B3 human lens epithelial cells. Dual-luciferase reporter assay and Western blot showed that miR-326 inhibited FGF1 expression by directly targeting its mRNA 3'-UTR. Consistent with this result, miR-326 antagomir enhanced FGF1 protein level. In addition to FGF1, miR-326 antagomir also enhanced ßB2 expression and this enhancement was abolished by transfection of HLEC-B3 cells with FGF1 shRNA. These data demonstrated that miR-326 antagomir increased ßB2 expression via upregulating FGF1, which was further confirmed by the studies in a rat model of selenite-induced cataract. This work suggests that miR-326 antagomir might be a promising candidate to prevent progression of age-related cataract.


Subject(s)
Antagomirs/metabolism , Cataract/metabolism , Fibroblast Growth Factor 1/metabolism , MicroRNAs/antagonists & inhibitors , beta-Crystallin B Chain/metabolism , 3' Untranslated Regions , Age Factors , Animals , Apoptosis , Cataract/genetics , Cataract/pathology , Cataract/therapy , Cell Line , Disease Progression , Epithelial Cells/cytology , Fibroblast Growth Factor 1/genetics , Humans , Lens, Crystalline/cytology , Rats, Sprague-Dawley , Up-Regulation
4.
Tumour Biol ; 37(11): 14885-14894, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27644246

ABSTRACT

γ-Aminobutyric acid (GABA) is a natural non-protein amino acid, which broadly exists in many plant parts and is widely used as an ingredient in the food industry. In mammals, it is widely distributed in central nervous system and non-neural tissues. In addition to a primary inhibitory neurotransmitter in the central nervous system, endogenous GABA content has been found to be elevated in neoplastic tissues in colon cancer. However, the effect of extraneous GABA on colon cancer has rarely been reported. In this study, we found the inhibitory effects of GABA on the proliferation of colon cancer cells (CCCs). The amino acid also suppressed metastasis of SW480 and SW620 cells. To further study the correlated mechanism, we analyzed the changes in cell cycle distribution and found that GABA suppressed cell cycle progression through G2/M or G1/S phase. Furthermore, RNA sequencing analysis revealed GABA-induced changes in the mRNA expression of 30 genes, including EGR1, MAPK4, NR4A1, Fos, and FosB, in all the three types of CCC. Importantly, GABA enhanced the anti-tumor efficacy of oxaliplatin (OXA) in subcutaneous xenograft tumor model in nude mice. The data suggest that GABA inhibits colon cancer cell proliferation perhaps by attenuating EGR1-NR4A1 axis, EGR1-Fos axis, and by disrupting MEK-EGR1 signaling pathway. This work reveals the pharmacological value of GABA derived from food and suggests that exogenous GABA might play an auxiliary role in polychemotherapy of colon cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Organoplatinum Compounds/pharmacology , gamma-Aminobutyric Acid/pharmacology , Animals , Base Sequence , Cell Line, Tumor , Drug Synergism , Early Growth Response Protein 1/metabolism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Oxaliplatin , Proto-Oncogene Proteins c-fos/metabolism , Sequence Analysis, RNA , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
Cell Discov ; 1: 15002, 2015.
Article in English | MEDLINE | ID: mdl-27462404

ABSTRACT

In addition to terminating neurotransmission by hydrolyzing acetylcholine, synaptic acetylcholinesterase (AChES) has been found to have a pro-apoptotic role. However, the underlying mechanism has rarely been investigated. Here, we report a nuclear translocation-dependent role for AChES as an apoptotic deoxyribonuclease (DNase). AChES polypeptide binds to and cleaves naked DNA at physiological pH in a Ca(2+)-Mg(2+)-dependent manner. It also cleaves chromosomal DNA both in pre-fixed and in apoptotic cells. In the presence of a pan-caspase inhibitor, the cleavage still occurred after nuclear translocation of AChES, implying that AChES-DNase acts in a CAD- and EndoG-independent manner. AChE gene knockout impairs apoptotic DNA cleavage; this impairment is rescued by overexpression of the wild-type but not (aa 32-138)-deleted AChES. Furthermore, in comparison with the nuclear-localized wild-type AChES, (aa 32-138)-deleted AChES loses the capacity to initiate apoptosis. These observations confirm that AChES mediates apoptosis via its DNase activity.

6.
J Mol Cell Biol ; 6(3): 231-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24748645

ABSTRACT

Thymine DNA glycosylase (TDG), an enzyme that initiates the repair of G/T and G/U mismatches, has been lately found crucial in embryonic development to maintain epigenetic stability and facilitate the active DNA demethylation. Here we report a novel role of TDG in Wnt signaling as a transcriptional coactivator of ß-catenin/TCFs complex. Our data show that TDG binds to the transcriptional factor family LEF1/TCFs and potentiates ß-catenin/TCFs transactivation, while TDG depletion suppresses Wnt3a-stimulated reporter activity or target gene transcription. Next, we show that CBP, a known coactivator, is also required for TDG function through forming a cooperative complex on target promoters. Moreover, there is an elevation of TDG levels in human colon cancer tissue, and knockdown of TDG inhibits proliferation of the colon cells. Overall, our results reveal that TDG, as a new coactivator, promotes ß-catenin/TCFs transactivation and functionally cooperates with CBP in canonical Wnt signaling.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/genetics , Peptide Fragments/metabolism , Sialoglycoproteins/metabolism , Thymine DNA Glycosylase/metabolism , Transcriptional Activation/genetics , beta Catenin/genetics , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HEK293 Cells , HeLa Cells , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Up-Regulation/genetics , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
7.
J Mol Cell Biol ; 3(4): 250-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21377978

ABSTRACT

Acetylcholinesterase (AChE) is emerging as an important contributor to apoptosis in various cell types. However, overexpression of AChE does not initiate apoptosis, and cells which express AChE at basal levels grow normally, suggesting that AChE may function differently between normal and apoptotic conditions. In this study, we determined that an AChE-derived protein (∼55 kDa) positively correlated with cellular apoptotic levels. The 55 kDa AChE protein was not a result of a novel splice variant of the AChE primary transcript. Instead, it was determined to be a cleaved fragment of the full-length 68 kDa AChE protein that could not be inhibited by cycloheximide (CHX) but could be suppressed by caspase inhibitors in apoptotic PC-12 cells. Furthermore, activation of the Akt cascade abolished the 55 kDa protein, and both AChE protein forms (68 and 55 kDa) accumulated in the nucleus during apoptosis. In a mouse model for ischemia/reperfusion (I/R)-induced acute renal failure, the 55 kDa AChE protein was detected in the impaired organs but not in the normal ones, and its levels correlated with the genotype of the mice. In summary, a 55 kDa AChE protein resulting from the cleavage of 68 kDa AChE is induced during apoptosis, and it is negatively regulated by the Akt pathway. This study suggests that an alternative form of AChE may play a role in apoptosis.


Subject(s)
Acetylcholinesterase/metabolism , Apoptosis , Gene Expression Regulation, Enzymologic , Proto-Oncogene Proteins c-akt/metabolism , Acetylcholinesterase/chemistry , Acetylcholinesterase/genetics , Animals , Cell Line , Cycloheximide/pharmacology , Disease Models, Animal , Enzyme Activation , Humans , Mice , RNA Interference , RNA, Small Interfering/metabolism , Rats , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reperfusion Injury/metabolism , Signal Transduction
8.
Bioorg Med Chem ; 14(7): 2438-45, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16326105

ABSTRACT

Previously, we found that 3,4-(methylenedioxy)-1-(2',3'-epoxypropyl)-benzene (safrole oxide) induced a typical apoptosis in A549 human lung cancer cells by activating caspase-3, -8, and -9. In this study, we further investigated which upstream pathways were activated by safrole oxide during the apoptosis. Immunofluorescence assay combined with laser scanning confocal microscopy revealed that both Fas and Fas ligand (FasL) were up-regulated by the small molecule. In addition, Fas protein distribution was altered, showing a clustering distribution instead of a homogeneous one. Subsequently, Western blot analysis confirmed the up-regulations of Fas and its membrane-binding form of FasL (m-FasL), as well as P53 protein. Conversely, safrole oxide hardly affected integrin beta4 subunit expression or distribution, which was reflected from the data obtained by immunofluorescence assay combined with laser scanning confocal microscopy. The results suggested that Fas/FasL pathway might be involved in safrole oxide-induced apoptosis of A549 cells, while integrin beta4 might be irrelevant to the apoptosis. Nevertheless, we first found the strong expression of integrin beta4 in A549 cells. The study first suggested that safrole oxide might be used as a small molecular promoter of Fas/FasL pathway to elicit apoptosis in A549 cells, which would lay the foundation for us to insight into the new strategies for lung cancer therapy.


Subject(s)
Apoptosis/drug effects , Integrin beta4/metabolism , Lung Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Safrole/analogs & derivatives , Tumor Necrosis Factors/metabolism , Cell Line, Tumor , Fas Ligand Protein , Humans , Integrin beta4/drug effects , Membrane Glycoproteins/drug effects , Receptors, Tumor Necrosis Factor/drug effects , Safrole/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , fas Receptor
9.
Bioorg Med Chem Lett ; 16(1): 81-3, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16242324

ABSTRACT

Previously we found that 3,4-(methylenedioxy)-1-(2',3'-epoxypropyl)-benzene (safrole oxide) induced a typical apoptosis in A549 human lung cancer cells. In this study, we further investigated which caspases were activated by safrole oxide during the apoptosis. The data showed that the activity of caspase-3, -8, and -9 was significantly enhanced by the compound, which suggested that safrole oxide might be used as a caspase promoter to initiate lung cancer cell apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Caspases/metabolism , Chemistry, Pharmaceutical/methods , Lung Neoplasms/drug therapy , Safrole/analogs & derivatives , Antineoplastic Agents/chemistry , Caspase 3 , Caspase 6 , Caspase 8 , Caspase 9 , Cell Line, Tumor , Colorimetry , Drug Design , Humans , Inflammation , Lung Neoplasms/pathology , Models, Chemical , Safrole/pharmacology , Time Factors , Up-Regulation
10.
Bioorg Med Chem ; 13(13): 4176-83, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15886004

ABSTRACT

A novel small molecule, 1-ethoxy-3-(3,4-methylenedioxyphenyl)-2-propanol (EOD), was synthesized in our laboratory. Previously, we reported pharmacological properties of EOD, triggering apoptosis in Human umbilical vein endothelial cells (HUVECs). Here, we further investigated the effects of EOD on the growth of A549 human lung cancer cells. EOD treatment induced apoptosis in A549 cells via up-regulating the expression of P53 protein, blocking cell cycle partly at G1 phase, and ultimately activating caspase-3. In contrast, caspase-8 might be irrelevant to EOD-triggered apoptosis. This study indicated that EOD might be a potential chemopreventive agent for lung cancer. The work would encourage us to add more novel compounds to our 'library' of small molecules derived through modern synthetic organic chemistry, and would drive us to determine the proteins that the compounds target.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/drug effects , Lung Neoplasms/drug therapy , Safrole/analogs & derivatives , Umbilical Veins/drug effects , Caspase 3 , Caspase 8 , Caspases/metabolism , Chromatin/metabolism , Endothelial Cells/cytology , G1 Phase/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Propanols , Safrole/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Umbilical Veins/cytology
11.
Exp Lung Res ; 30(6): 419-29, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15524402

ABSTRACT

3,4-(Methylenedioxy)-1-(2',3'-epoxypropyl)-benzene (safrole oxide) was synthesized in the authors' laboratory. To investigate the effects of safrole oxide on the growth and apoptosis of A549 human lung cancer cells, the authors treated the cells with safrole oxide, 112.36 to 449.44 micromol/L, for 24 to 48 hours. The results showed that the drug led A549 cells to apoptosis and blocked cell cycle completely at G1 phase and partly at G(2)-M phase. To further study the correlated mechanism, the authors examined P53 and H-Ras protein expressions by using immunofluorescence assay. They found that the expression of P53 was dramatically up-regulated but the expression of H-Ras was hardly affected by safrole oxide, 224.72 micromol/L, within 24 hours. Taken together, these results revealed that safrole oxide could induce apoptosis of A549 cells and suggested that safrole oxide might perform its function by blocking cells completely at G1 phase and partly at G(2)-M phase, and also by up-regulating the expression of P53 protein. These findings would raise exciting possibilities for cancer therapy in future.


Subject(s)
Apoptosis/drug effects , Lung Neoplasms/pathology , Safrole/analogs & derivatives , Safrole/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Humans , L-Lactate Dehydrogenase/metabolism , Lung Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism
12.
Endothelium ; 11(5-6): 267-73, 2004.
Article in English | MEDLINE | ID: mdl-15763947

ABSTRACT

Two safrole oxide derivatives, 1-propoxy-3-(3,4-methylenedioxyphenyl)-2-propanol (FOD) and 1-isopropoxy-3-(3,4-methylenedioxyphenyl)-2-propanol (GOD), were newly synthesized as promoters of apoptosis in vascular endothelial cells. The purpose of this study was to investigate the effects of these two safrole oxide derivatives on cell growth and apoptosis induced by deprivation of survival factors (serum and fibroblast growth factors, aFGF and bFGF) in vascular endothelial cells (VECs). MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium) method, agarose gel electrophoresis, laser scanning confocal microscopy, flow cytometry (FCM), and immunofluorescence assay were used. The cells deprived of FGF and serum were exposed to FOD or GOD 30 to 90 mg x L(-1) for 24 h, cell growth was suppressed (p < .05), whereas detachment and DNA fragmentation of these cells were promoted (p < .01). When the cells were treated with FOD 90 mg x L(-) for 24 h, apoptosis rate was 14.99% (p < .01). There were more cells in G2-M phase and less cells in S phase. At 90 mg x L(-1) concentration, GOD blocked 77.03%of the cells at G0-G1 phase., P53 level in VEC exposed to FOD or GOD was increased (p < .01). The data suggested that FOD and GOD might promote apoptosis of VEC by affect the cell cycle distribution, whereas P53 was involved in this pathway.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/drug effects , Propanols/pharmacology , Cell Cycle/drug effects , Cell Nucleus/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Endothelial Cells/cytology , Endothelium, Vascular/drug effects , Gene Expression/drug effects , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Vascul Pharmacol ; 40(3): 183-7, 2003 Oct.
Article in English | MEDLINE | ID: mdl-13678651

ABSTRACT

Two safrole oxide derivatives, 1-methoxy-3-(3,4-methylenedioxyphenyl)-2-propanol (MOD) and 1-ethoxy-3-(3,4-methylenedioxyphenyl)-2-propanol (EOD), were newly synthesized as promoters of apoptosis in vascular endothelial cells (VECs). The purpose of this study was to investigate the effects of these two safrole oxide derivatives on cell growth and apoptosis induced by deprivation of survival factors (serum and fibroblast growth factors, aFGF and bFGF) in VECs. Morphological changes were observed with light microscopy. Cell growth was determined by using MTT (3-[4, 5-dimethyl thiazol-2-yl]-2, 5-diphenytetrazolium) method. DNA fragmentation was analyzed by agarose gel electrophoresis and fluorescence microscopy. Apoptosis rate and cell cycle distribution were analyzed by flow cytometry (FCM). The cells deprived of FGF and serum were exposed to MOD 10-40 mg l(-1) for 24 h. Cell growth was suppressed (P<.01), while detachment and DNA fragmentation of these cells were promoted (P<.01). When the cells were treated with MOD30 mg l(-1) for 24 h, apoptosis rate was 21.43% (P<.01). The fact that 66.50% of the cells were trapped in S phase of cell cycle indicated that the cell cycle was blocked at S phase. Treated with EOD 10-40 mg l(-1) for 24 h, the cells were observed; the results showed that VEC growth was inhibited and the apoptosis was triggered (P<.01). At 30 mg l(-1) concentration, EOD blocked 55.22% of the cells at S phase. The data suggested that MOD and EOD might promote apoptosis of VEC by blocking the cell cycle at S phase.


Subject(s)
Apoptosis/drug effects , Endothelium, Vascular/drug effects , Safrole/analogs & derivatives , Safrole/pharmacology , Cell Division/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , DNA Fragmentation/drug effects , Endothelium, Vascular/cytology , Fibroblast Growth Factors/pharmacology , Flow Cytometry , Humans , Interphase/drug effects , Safrole/chemical synthesis , Serum/chemistry , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...