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










Publication year range
1.
Environ Sci Technol ; 57(49): 20460-20469, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38019752

ABSTRACT

Biodiesel, derived from alkyl esters of vegetable oils or animal fats, has gained prominence as a greener alternative to diesel due to its reduced particle mass. However, it remains debatable whether biodiesel exposure has more severe health issues than diesel. This study performed high-resolution mass spectrometry to examine the detailed particle chemical compositions and lipidomics analysis of human lung epithelial cells treated with emissions from biodiesel and diesel fuels. Results show the presence of the peak substances of CHO compounds in biodiesel combustion that contain a phthalate ester (PAEs) structure (e.g., n-amyl isoamyl phthalate and diisobutyl phthalate). PAEs have emerged as persistent organic pollutants across various environmental media and are known to possess endocrine-disrupting properties in the environment. We further observed that biodiesel prevents triglyceride storage compared to diesel and inhibits triglycerides from becoming phospholipids, particularly with increased phosphatidylglycerols (PGs) and phosphatidylethanolamines (PEs), which potentially could lead to a higher probability of cancer metastasis.


Subject(s)
Air Pollutants , Vehicle Emissions , Animals , Humans , Vehicle Emissions/analysis , Biofuels/analysis , Lipid Metabolism , Gasoline/analysis , Air Pollutants/analysis
2.
Biochem Pharmacol ; 210: 115462, 2023 04.
Article in English | MEDLINE | ID: mdl-36849061

ABSTRACT

Identifying the key factors mediating the progression from hypertension to cardiac hypertrophy is critically important for developing a strategy to protect against heart failure. Serum exosomes have been revealed to be involved in the development of cardiovascular disease. In the current study, we found that either serum or serum exosomes derived from SHR induced hypertrophy in H9c2 cardiomyocytes. SHR Exo injection through the tail vein for 8 weeks induced left ventricular wall thickening and decreased cardiac function in C57BL/6 mice. SHR Exo carried the renin-angiotensin system (RAS) proteins AGT, renin, and ACE into cardiomyocytes, which increased the autocrine secretion of Ang II. Moreover, the AT1-type receptor antagonist telmisartan prevented hypertrophy of H9c2 cells induced by SHR Exo.These results identified a novel role of exosomes derived from SHR serum in cardiac hypertrophy and revealed that SHR Exo induced cardiac hypertrophy by carrying AGT, renin, and ACE proteins into cardiomyocytes to increase their autocrine secretion of Ang II. The emergence of this new mechanism will help us better understand how hypertension progresses to cardiac hypertrophy.


Subject(s)
Exosomes , Hypertension , Rats , Mice , Animals , Angiotensin II/metabolism , Rats, Inbred SHR , Myocytes, Cardiac/metabolism , Renin/metabolism , Exosomes/metabolism , Mice, Inbred C57BL , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Renin-Angiotensin System , Hypertension/metabolism
3.
Biochem J ; 479(5): 629-640, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35175311

ABSTRACT

Iodide (I-) is crucial to thyroid function, and its regulation in thyrocytes involves ion transporters and reactive oxygen species (ROS). However, the extent of 2Cl-/H+ exchanger (ClC-3) involvement in the iodide (I-) efflux from thyrocytes remains unclear. Therefore, we examined the effects of ClC-3 on I- efflux. ClC-3 expression was found to significantly alter the serum TT3 and TT4 concentrations in mice. We further found that excess I- stimulation affected ClC-3 expression, distribution, and I- efflux in FRTL-5 cells. Immunofluorescence analyses indicated that ClC-3 mainly accumulated in the cell membrane and co-localized with ß-tubulins after 24 h of excess I- treatment, and that this process depended on ROS production. Thus, ClC-3 may be involved in I- efflux at the apical pole of thyrocytes via excess I--induced ROS production and ß-tubulin polymerization. Our results reveal novel insights into the role of ClC-3 in I- transport and thyroid function.


Subject(s)
Chloride Channels/metabolism , Thyroid Epithelial Cells , Animals , Biological Transport , Iodides , Mice , Protons , Reactive Oxygen Species , Tubulin
4.
Anticancer Agents Med Chem ; 22(6): 1102-1110, 2022.
Article in English | MEDLINE | ID: mdl-34353269

ABSTRACT

BACKGROUND: Despite emerging research on new treatment strategies, chemotherapy remains one of the most important therapeutic modalities for cancers. Imidazopyridines are important targets in organic chemistry and, given their numerous applications, they are worthy of attention. OBJECTIVE: The objective of this study was to design and synthesize a novel series of imidazo[1,2-a]pyridine-derived compounds and investigate their antitumor effects and the underlying mechanisms. METHODS: Imidazo[1,2-a]pyridine-derived compounds were synthesized with new strategies and conventional methods. The antitumor activities of the new compounds were evaluated by MTT assay. Flow cytometry and immunofluorescence were performed to examine the effects of the most effective antiproliferative compound on cell apoptosis. Western blot analysis was used to assess the expression of apoptotic proteins. RESULTS: Fifty-two new imidazo[1,2-a]pyridine compounds were designed and successfully synthesized. The compound, 1-(imidazo[1,2-a]pyridin-3-yl)-2-(naphthalen-2-yl)ethane-1,2-dione, named La23, showed high potential for suppressing the viability of HeLa cells (IC50 15.32 µM). La23 inhibited cell proliferation by inducing cell apoptosis, and it reduced the mitochondrial membrane potential of HeLa cells. Moreover, treatment with La23 appeared to increase the expression of apoptotic-related protein P53, Bax, cleaved caspase-3, and cytochrome c at a low concentration range. CONCLUSION: The novel imidazo[1,2-a]pyridine compound, La23, was synthesized and it suppressed cell growth by inducing cell apoptosis via the p53/Bax mitochondrial apoptotic pathway.


Subject(s)
Antineoplastic Agents , Tumor Suppressor Protein p53 , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , Cell Survival , HeLa Cells , Humans , Membrane Potential, Mitochondrial , Pyridines , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
5.
J Leukoc Biol ; 110(6): 1081-1090, 2021 12.
Article in English | MEDLINE | ID: mdl-34779530

ABSTRACT

T cells play an important role in antitumor immunity. Numbers and function of T cells are controlled by regulating the uptake and utilization of nutrients, and their antitumor activity can be promoted by targeting metabolic pathways. In this review, we highlight the relationship between metabolism and cellular function of T cells. Specifically, we emphasize the metabolic state of tumor-infiltrating T cells and review key pathways that affect the antitumor function of T cells. In the field of tumor immunotherapy, targeting T cell metabolism to enhance the immune response is a new therapeutic strategy for enhancing immunotherapy combined with traditional treatments.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/therapy
6.
Biochem Pharmacol ; 193: 114791, 2021 11.
Article in English | MEDLINE | ID: mdl-34582774

ABSTRACT

The representative morphological features of pyroptosis are excessive cell swelling and subsequent membrane rupture. However, the mechanism underlying the cell's inherent inability to regulate volume during the progression of pyroptosis is poorly understood. In the current study, we found that both volume-activated chloride currents (Icl, vol) and the regulatory volume decrease (RVD) were markedly decreased in bone marrow-derived macrophages (BMDMs) undergoing pyroptosis induced by lipopolysaccharides (LPS) and nigericin. The inhibition of ICl, vol and RVD by the chloride channel blockers, tamoxifen or DCPIB, led to the emergence of pyroptosis-like phenotypes such as activated-caspase-1, pyroptotic-body-like bubbles, and a fried-egg-like appearance. Moreover, the expression of the volume-activated chloride channel (VRAC) constituent protein Leucine-Rich Repeat-Containing 8A (LRRC8A) was significantly down-regulated in pyroptotic BMDMs treated with LPS and nigericin. The silencing of LRRC8A expression by small interfering RNA (si)-LRRC8A transfection not only reduced ICl, vol and RVD, but also caused BMDMs to show pyroptosis-like manifestations such as activated-caspase-1, membrane bubbles, and have a fried-egg-like appearance. These results reveal a new mechanism for the loss of volume regulation in the process of pyroptotic cell swelling and strongly suggest that a functional deficiency of VRAC/LRRC8A plays a key role in this disorder.


Subject(s)
Chloride Channels/metabolism , Lipopolysaccharides/toxicity , Nigericin/toxicity , Pyroptosis/drug effects , Animals , Anti-Bacterial Agents/toxicity , Biomarkers , Cyclopentanes/pharmacology , Estrogen Antagonists/pharmacology , Gene Expression Regulation/drug effects , Indans/pharmacology , Macrophages , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , RNA, Small Interfering , Tamoxifen/pharmacology
7.
Biotechnol Lett ; 43(8): 1675-1684, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34014413

ABSTRACT

Wound exudate holds great clinical and research potential in wound repair via paracrine signaling. In essence, exudate is modified serum that contains a high concentration of exosomes. The aim of this study was to investigate the role of serum-derived exosomes in scald wound healing of NIH mice skin and to explore the underlying mechanisms. Hence, we constructed a deep second-degree scald model in NIH mice, testing the benefits of exosomes in the scald wound healing. The scratch wound assay, apoptosis assay and MTT assay were conducted to assess the effects of serum-derived exosomes on migration, apoptosis and proliferation of HaCaT cells and fibroblasts. Our results showed that serum-derived exosomes injected subcutaneously entered cells and effectively accelerated wound healing processes in mice. Additionally, serum-derived exosomes optimized functions of cells related to skin injury repair by stimulating fibroblast proliferation, promoting HaCaT cell migration, and suppressing apoptosis of HaCaT cells induced by heat stress. Further study revealed that serum-derived exosomes enhanced phosphorylation of the serine-threonine kinase Akt in scalded skin tissue. These results suggest a potential clinical use of serum-derived exosomes for treating skin scald.


Subject(s)
Exosomes , Proto-Oncogene Proteins c-akt/metabolism , Wound Healing/drug effects , Animals , Biological Products/pharmacology , Cell Line , Male , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/chemistry , Rats, Sprague-Dawley , Serum/chemistry , Skin/cytology , Skin/drug effects , Skin/injuries
8.
Mitochondrial DNA B Resour ; 6(1): 60-61, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33521267

ABSTRACT

Meclicope pteleifolia is a traditional medicinal herb and edible plant in Southeast China. Here, we report the complete chloroplast genome of M. pteleifolia. The chloroplast genome is 159,012 bp in length with 38.33% GC content, containing a small single-copy (SSC) region (18,609 bp), a large single-copy (LSC) region (851 bp), and a pair of inverted repeats (IRs: 27,640 bp each). A total of 131 genes were predicted, including 84 protein-coding genes, 8 ribosomal RNA genes, 37 tRNA genes, and 2 pseudogenes. Phylogenetic analysis based on chloroplast genomes of 17 plant species shows that M. pteleifolia is closest to Zanthoxylum and Casimiroa. These complete chloroplast genomes can be subsequently used for researches of Rutaceae.

9.
Int J Dev Biol ; 64(10-11-12): 471-477, 2020.
Article in English | MEDLINE | ID: mdl-33336709

ABSTRACT

FGF signaling pathway is imperative for definitive endoderm (DE) differentiation from human embryonic stem cells (hESCs), which always accompanies an epithelial-to-mesenchymal transition (EMT) process. However, whether there is an association between FGF signaling and the EMT during DE formation in vitro has remained elusive. In the present study, we identify that several FGF family members were significantly activated during the differentiation of hESCs toward DE. Inhibition of FGF signaling by an efficient and selective inhibitor BGJ398 abolishes both the EMT and DE induction by blocking the activation of the zinc-finger transcription factor SNAI1 which is a direct transcriptional repressor of cell adhesion protein CDH1. In addition, cell proliferation is also severely influenced by attenuating the FGF signaling. Collectively, we propose that the FGF signaling promotes the DE formation through mediating the EMT and cell proliferation.


Subject(s)
Endoderm/cytology , Epithelial-Mesenchymal Transition , Fibroblast Growth Factors/physiology , Signal Transduction , Cell Differentiation , Cell Proliferation , Ectoderm/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Fibroblast Growth Factors/antagonists & inhibitors , Gene Expression , Humans , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology
10.
Am J Transl Res ; 12(11): 7420-7429, 2020.
Article in English | MEDLINE | ID: mdl-33312378

ABSTRACT

Fibroblasts migrating to peritoneum injuries play an important role in the development of postoperative peritoneal adhesions due to the excessive synthesis and deposition of extracellular matrix (ECM). This effect is mainly induced by the transforming growth factor-ß (TGF-ß). Studies indicate that elevated TGF-ß1 levels and TGF-ß1/Smad signaling are both implicated in the formation of peritoneal adhesions. To confirm the effect of TGF-ß1/Smad signaling interference in regulating excessive ECM synthesis, a total of four different R-Smad-targeting small interference RNA (siRNA) duplexes (Smad2-500, Smad2-956, Smad3-378, Smad3-1385) were tested in this study using a TGF-ß1-stimulated adhesion tissue fibroblasts (ATFs) cell model. The in vitro assessments show that all proposed siRNAs are capable of significantly downregulating the mRNA and protein levels of Smad2 and Smad3 in ATFs. They also inhibit the phosphorylation of both Smads, which confirms their effect in blocking the TGF-ß1/Smad signaling pathway. Moreover, the siRNA duplexes can appreciably lower the elevated levels of fibronectin and collagen 3 alpha 1 (COL3A1) in TGF-ß1-stimulated ATFs, and the Smad3-378 siRNA can actually restore both molecules (fibronectin and COL3A1) to normal levels. Therefore, the Smad3-378 siRNA is suitable for both adhesion prevention and wound healing. Overall, our results indicate that postoperative adhesion prophylaxis may be achieved by temporarily blocking TGF-ß1/Smad signaling transduction.

11.
In Vitro Cell Dev Biol Anim ; 56(5): 378-385, 2020 May.
Article in English | MEDLINE | ID: mdl-32514718

ABSTRACT

Whether mitochondrial remodeling and metabolic reprogramming occur during the differentiation of human embryonic stem cells (hESCs) to definitive endoderm (DE) is unknown. We found that fragmented and punctate mitochondria in undifferentiated hESCs progressively fused into an extensive and branched network upon DE differentiation. Mitochondrial mass and mitochondrial DNA (mtDNA) content were significantly increased with the upregulated expression of mitochondrial biogenesis regulator PGC1-A upon DE differentiation, accompanied by the rise of the amount of ATP (2.5-fold) and its by-product reactive oxygen species (2.0-fold). We observed that in contrast to a shutoff of glycolysis, expressions of oxidative phosphorylation (OXPHOS) genes were increased, indicating that a transition from glycolysis to OXPHOS was tightly coupled to DE differentiation. In the meantime, we discovered that inhibition of TGF-ß signaling led to impaired mitochondrial biogenesis and disturbed metabolic switch upon DE differentiation. Our work, for the first time, reports that TGF-ß signaling-dependent mitochondrial biogenesis and metabolic reprogramming occur during early endodermal differentiation.


Subject(s)
Cell Differentiation , Endoderm/cytology , Organelle Biogenesis , Transforming Growth Factor beta/metabolism , Cell Differentiation/drug effects , DNA, Mitochondrial/genetics , Gene Dosage , Glycolysis/drug effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects
12.
Histochem Cell Biol ; 153(5): 307-321, 2020 May.
Article in English | MEDLINE | ID: mdl-32078038

ABSTRACT

Serine-52 (Ser52) is the major physiologic site of keratin 18 (K18) phosphorylation. Here, we report that serine-52 phosphorylated K18 (phospho-Ser52 K18) accumulated on centrosomes in a cell cycle-dependent manner. Moreover, we found that phospho-Ser52 K18 was located at the proximal end of the mother centriole. Transfection with the K18 Ser52 → Ala (K18 S52A) mutant prevented centriole localization of phospho-Ser52 K18 and resulted in separation of the mother-daughter centrioles. Inhibition of microtubule polymerization led to the disappearance of aggregated phospho-Ser52 K18 on the centrosome; removal of inhibitors resulted in reaccumulation of phospho-Ser52 K18 in microtubule-organizing centers. Transfection with a K18 S52A mutant inhibited microtubule nucleation. These results reveal a cell cycle-dependent change in centrosome localization of phospho-Ser52 k18 and strongly suggest that the phosphorylation status of Ser52 K18 of mother centrioles plays a critical role in maintaining a tight engagement between mother and daughter centrioles and also contributes to microtubule nucleation.


Subject(s)
Cell Cycle , Centrioles/metabolism , Keratin-18/metabolism , Microtubules/metabolism , Serine/metabolism , Animals , Cells, Cultured , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation
13.
Naunyn Schmiedebergs Arch Pharmacol ; 393(3): 501-510, 2020 03.
Article in English | MEDLINE | ID: mdl-31659404

ABSTRACT

5-Nitro-2-(3-phenylpropylamino)-benzoic acid (NPPB) is a non-specific chloride channel blocker. Peritoneal adhesion is an inevitable complication of abdominal surgery and remains an important clinical problem, leading to chronic pain, intestinal obstruction, and female infertility. The aim of this study is to observe the effects of NPPB on peritoneal adhesions and uncover the underlying mechanism. The formation of postoperative peritoneal adhesions was induced by mechanical injury to the peritoneum of rats. MTT assay and wound-healing assay were used to evaluate proliferation and migration of primary cultured adhesion fibroblasts (AFB) respectively. Whole-cell chloride currents were measured using a fully automated patch-clamp workstation. Cell volume changes were monitored by light microscopy and video imaging. Our results demonstrated that NPPB could significantly prevent the formation of peritoneal adhesion in rats and inhibit the proliferation of AFB in a concentration-dependent manner. NPPB also reduced the migration of AFB cells with an IC50 of 53.09 µM. A 47% hypotonic solution successfully activated the ICl,vol in AFB cells. The current could be blocked by extracellular treatment with NPPB. Moreover, 100 µM NPPB almost completely eliminated the capacity of regulatory volume decrease (RVD) in these cells. These data indicate that NPPB could prevent the formation of postoperative peritoneal adhesions. The possible mechanism may be through the inhibition of the proliferation and migration of AFB cells by modulating ICl,vol and cell volume. These results suggest a potential clinical use of NPPB for preventing the formation of peritoneal adhesions.


Subject(s)
Cell Movement/drug effects , Chloride Channels/antagonists & inhibitors , Nitrobenzoates/therapeutic use , Peritoneum/drug effects , Postoperative Complications/drug therapy , Tissue Adhesions/drug therapy , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Movement/physiology , Cells, Cultured , Chloride Channels/physiology , Fibroblasts/drug effects , Fibroblasts/physiology , Nitrobenzoates/pharmacology , Peritoneum/physiopathology , Postoperative Complications/etiology , Postoperative Complications/physiopathology , Rats , Rats, Sprague-Dawley , Tissue Adhesions/etiology , Tissue Adhesions/physiopathology
14.
J Cell Physiol ; 234(5): 6611-6623, 2019 05.
Article in English | MEDLINE | ID: mdl-30230544

ABSTRACT

Chloride channel-3 (ClC-3), a member of the ClC family of voltage-gated Cl- channels, is involved in the resistance of tumor cells to chemotherapeutic drugs. Here, we report a new mechanism for ClC-3 in mediating multidrug resistance (MDR). ClC-3 was highly expressed in the P-glycoprotein (P-gp)-dependent human lung adenocarcinoma cell line (A549)/paclitaxel (PTX) and the human breast carcinoma cell line (MCF-7)/doxorubicin (DOX) resistant cells. Changes in the ClC-3 expression resulted in the development of drug resistance in formerly drug-sensitive A549 or MCF-7 cells, and drug sensitivity in formerly drug-resistant A549/Taxol and MCF-7/DOX cells. Double transgenic MMTV-PyMT/CLCN3 mice with spontaneous mammary cancer and ClC-3 overexpression demonstrated drug resistance to PTX and DOX. ClC-3 expression upregulated the expression of MDR1 messenger RNA and P-gp by activating the nuclear factor-κB (NF-κB)-signaling pathway. These data suggest that ClC-3 expression in cancer cells induces MDR by upregulating NF-κB-signaling-dependent P-gp expression involving another new mechanism for ClC-3 in the development of drug resistance of cancers.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Breast Neoplasms/metabolism , Chloride Channels/metabolism , Drug Resistance, Neoplasm/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Multiple/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , NF-kappa B/metabolism , Transcriptional Activation/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
15.
Gene ; 642: 335-342, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29158167

ABSTRACT

Isoprenaline, an activator of ß-adrenergic receptor, has been found to induce cardiac hypertrophy in vivo and in vitro, but the exact mechanism is still unclear. ClC-3 is a member of the chloride channel family and is highly expressed in mammalian myocardium. In the present study, the role of ClC-3 in isopronaline-induced cardiac hypertrophy was investigated. We found that ClC-3 expression was reduced in isoprenaline-induced hypertrophic H9c2 cells, primary rat neonatal cardiomyocytes and myocardium of C57/BL/6 mice, and this reduction was prevented by the pretreatment of propranolol. Adeno-associated virus 9 (AAV9)-mediated ClC-3 expression in myocardium decreased heart mass index, thinned interventricular septum and left ventricular wall and lowered the mRNA expression of natriuretic peptide type A (ANF) and ß-myosin heavy chain (ß-MHC). Our results showed that ClC-3 played an important role in ß-adrenergic cardiac hypertrophy which could be associated with ANF and ß-MHC, and all these findings suggested that ClC-3 may be a novel therapeutic target for the prevention or treatment of myocardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Chloride Channels/metabolism , Isoproterenol/adverse effects , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cell Line , Chloride Channels/genetics , Dependovirus/genetics , Disease Models, Animal , Down-Regulation , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Propranolol/pharmacology , Rats , Receptors, Atrial Natriuretic Factor/genetics , Ventricular Myosins/genetics
16.
Sci Rep ; 6: 30276, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27451945

ABSTRACT

It was shown in this study that knockdown of ClC-3 expression by ClC-3 siRNA prevented the activation of hypotonicity-induced chloride currents, and arrested cells at the G0/G1 phase in nasopharyngeal carcinoma CNE-2Z cells. Reconstitution of ClC-3 expression with ClC-3 expression plasmids could rescue the cells from the cell cycle arrest caused by ClC-3 siRNA treatments. Transfection of cells with ClC-3 siRNA decreased the expression of cyclin D1, cyclin dependent kinase 4 and 6, and increased the expression of cyclin dependent kinase inhibitors (CDKIs), p21 and p27. Pretreatments of cells with p21 and p27 siRNAs depleted the inhibitory effects of ClC-3 siRNA on the expression of CDK4 and CDK6, but not on that of cyclin D1, indicating the requirement of p21 and p27 for the inhibitory effects of ClC-3 siRNA on CDK4 and CDK6 expression. ClC-3 siRNA inhibited cells to progress from the G1 phase to the S phase, but pretreatments of cells with p21 and p27 siRNAs abolished the inhibitory effects of ClC-3 siRNA on the cell cycle progress. Our data suggest that ClC-3 may regulate cell cycle transition between G0/G1 and S phases by up-regulation of the expression of CDK4 and CDK6 through suppression of p21 and p27 expression.


Subject(s)
Carcinoma/genetics , Chloride Channels/genetics , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Nasopharyngeal Neoplasms/genetics , Carcinoma/pathology , Cell Cycle/genetics , Cell Line, Tumor , Chloride Channels/antagonists & inhibitors , Cyclin D1/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression Regulation, Neoplastic , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , RNA, Small Interfering/genetics , S Phase/genetics , Transcriptional Activation/genetics
17.
Org Lett ; 18(15): 3582-5, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27388919

ABSTRACT

An efficient copper-catalyzed selective cross coupling of imidazo[1,2-a]pyridines with methyl hetarenes has been reported. This transformation opened a new route to synthesize the C-3 carbonyl imidazo[1,2-a]pyridine derivative, which is a common structural motif in natural products and pharmaceuticals. (18)O-labeling experiments indicated that the oxygen source of products originated from O2.

18.
Theranostics ; 6(2): 204-18, 2016.
Article in English | MEDLINE | ID: mdl-26877779

ABSTRACT

P-glycoprotein (P-gp) is encoded by the multidrug resistance (MDR1) gene and is well studied as a multi-drug resistance transporter. Peritoneal adhesion formation following abdominal surgery remains an important clinical problem. Here, we found that P-gp was highly expressed in human adhesion fibroblasts and promoted peritoneal adhesion formation in a rodent model. Knockdown of P-gp expression by intraperitoneal injection of MDR1-targeted siRNA significantly reduced both the peritoneal adhesion development rate and adhesion grades. Additionally, we found that operative injury up-regulated P-gp expression in peritoneal fibroblasts through the TGF-ß1/Smad signaling pathway and histone H3 acetylation. The overexpression of P-gp accelerated migration and proliferation of fibroblasts via volume-activated Cl(-) current and cell volume regulation by enhancing phosphorylation of the chloride channel-3. Therefore, P-gp plays a critical role in postoperative peritoneal adhesion formation and may be a valuable therapeutic target for preventing the formation of peritoneal adhesions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Chloride Channels/metabolism , Peritoneal Diseases/metabolism , Protein Processing, Post-Translational , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Cells, Cultured , Fibroblasts/metabolism , Gene Silencing , Humans , Laparotomy/adverse effects , Peritoneal Diseases/etiology , Phosphorylation , Postoperative Complications/metabolism , Rats , Up-Regulation
19.
J Pharmacol Sci ; 127(3): 332-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25837931

ABSTRACT

Pine needle oil from crude extract of pine needles has been used as an anti-cancer agent in Traditional Chinese Medicine. The α-pinene is a natural compound isolated from pine needle oil which has been shown anti-cancer activity. In previous study, we found that pine needle oil exhibited significant inhibitory effect on hepatoma carcinoma BEL-7402 cells. In this study, we investigate the inhibition of α-pinene on hepatoma carcinoma BEL-7402 cells in vitro and in vivo and further explore the mechanism. The results show that liver cancer cell growth was inhibited obviously with inhibitory rate of 79.3% in vitro and 69.1% in vivo, Chk1 and Chk2 levels were upregulated, CyclinB, CDC25 and CDK1 levels were downregulated.


Subject(s)
Antineoplastic Agents, Phytogenic , Carcinoma, Hepatocellular/pathology , G2 Phase Cell Cycle Checkpoints/genetics , Liver Neoplasms/pathology , M Phase Cell Cycle Checkpoints/genetics , Monoterpenes/pharmacology , Pinus/chemistry , Plant Oils/chemistry , Animals , Bicyclic Monoterpenes , CDC2 Protein Kinase , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Checkpoint Kinase 1 , Checkpoint Kinase 2/metabolism , Cyclin B/metabolism , Cyclin-Dependent Kinases/metabolism , Down-Regulation/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Mice, Nude , Monoterpenes/isolation & purification , Neoplasm Transplantation , Phytotherapy , Protein Kinases/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects
20.
Oncotarget ; 6(4): 2434-50, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25537517

ABSTRACT

The chloride channel-3 (ClC-3) protein is known to be a component of Cl- channels involved in cell volume regulation or acidification of intracellular vesicles. Here, we report that ClC-3 was highly expressed in the cytoplasm of metastatic carcinomatous cells and accelerated cell migration in vitro and tumor metastasis in vivo. High-grade expression of cytoplasmic ClC-3 predicted poor survival in cancer patients. We found that independent of its volume-activated Cl- channel properties, ClC-3 was able to promote cell membrane ruffling, required for tumor metastasis. ClC-3 mediated membrane ruffling by regulating keratin 18 phosphorylation to control ß1 Integrin recycling. Therefore, cytoplasmic ClC-3 plays an active and key role in tumor metastasis and may be a valuable prognostic biomarker and a therapeutic target to prevent tumor spread.


Subject(s)
Cell Membrane/metabolism , Chloride Channels/metabolism , Cytoplasm/metabolism , Neoplasms/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Chloride Channels/genetics , Cytoplasm/genetics , HeLa Cells , Humans , Immunoblotting , Integrin beta1/metabolism , Kaplan-Meier Estimate , Keratin-18/genetics , Keratin-18/metabolism , Mice, Transgenic , Microscopy, Confocal , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Prognosis , RNA Interference , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...