Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
J Cell Biol ; 223(4)2024 Apr 01.
Article En | MEDLINE | ID: mdl-38466167

Focal adhesions (FAs) are transmembrane protein assemblies mediating cell-matrix connection. Although protein liquid-liquid phase separation (LLPS) has been tied to the organization and dynamics of FAs, the underlying mechanisms remain unclear. Here, we experimentally tune the LLPS of PXN/Paxillin, an essential scaffold protein of FAs, by utilizing a light-inducible Cry2 system in different cell types. In addition to nucleating FA components, light-triggered PXN LLPS potently activates integrin signaling and subsequently accelerates cell spreading. In contrast to the homotypic interaction-driven LLPS of PXN in vitro, PXN condensates in cells are associated with the plasma membrane and modulated by actomyosin contraction and client proteins of FAs. Interestingly, non-specific weak intermolecular interactions synergize with specific molecular interactions to mediate the multicomponent condensation of PXN and are efficient in promoting FA assembly and integrin signaling. Thus, our data establish an active role of the PXN phase transition into a condensed membrane-associated compartment in promoting the assembly/maturation of FAs.


Focal Adhesions , Paxillin , Phase Separation , Humans , Actin Cytoskeleton , Focal Adhesions/metabolism , Integrins/metabolism , Paxillin/chemistry , Paxillin/metabolism
2.
EMBO Rep ; 24(11): e56850, 2023 11 06.
Article En | MEDLINE | ID: mdl-37846507

The remodeling and stiffening of the extracellular matrix (ECM) is a well-recognized modulator of breast cancer progression. How changes in the mechanical properties of the ECM are converted into biochemical signals that direct tumor cell migration and metastasis remain poorly characterized. Here, we describe a new role for the autophagy-inducing serine/threonine kinases ULK1 and ULK2 in mechanotransduction. We show that ULK1/2 activity inhibits the assembly of actin stress fibers and focal adhesions (FAs) and as a consequence impedes cell contraction and migration, independent of its role in autophagy. Mechanistically, we identify PXN/paxillin, a key component of the mechanotransducing machinery, as a direct binding partner and substrate of ULK1/2. ULK-mediated phosphorylation of PXN at S32 and S119 weakens homotypic interactions and liquid-liquid phase separation of PXN, impairing FA assembly, which in turn alters the mechanical properties of breast cancer cells and their response to mechanical stimuli. ULK1/2 and the well-characterized PXN regulator, FAK/Src, have opposing functions on mechanotransduction and compete for phosphorylation of adjacent serine and tyrosine residues. Taken together, our study reveals ULK1/2 as important regulator of PXN-dependent mechanotransduction.


Breast Neoplasms , Humans , Female , Paxillin/metabolism , Mechanotransduction, Cellular , Phosphorylation , Cell Movement , Serine/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
3.
Biochem Biophys Res Commun ; 477(4): 1031-1037, 2016 09 02.
Article En | MEDLINE | ID: mdl-27392714

Selective induction of cell death or growth inhibition of cancer cells is the future of chemotherapy. Clinical trials have found that cancer tissues are enriched with copper. Based on this finding, many copper-containing compounds and complexes have been designed to "copper" cancer cells using copper as bait. However, recent studies have demonstrated that copper boosts tumor development, and copper deprivation from serum was shown to effectively inhibit the promotion of cancer. Mechanistically, copper is an essential cofactor for mitogen-activated protein kinase (MAPK)/extracellular activating kinase (ERK) kinase (MEK), a central molecule in the BRAF/MEK/ERK pathway. Therefore, depleting copper from cancer cells by directly sequestering copper has a wider field for research and potential for combination therapy. Based on the affinity between sulfur and copper, we therefore designed sulfur nanoparticles (Nano-S) that detain copper, achieving tumor growth restriction. We found that spherical Nano-S could effectively bind copper and form a tighter surficial structure. Moreover, this Nano-S detention of copper effectively inhibited the proliferation of A375 melanoma and MCF-7 breast cancer cells with minimum toxicity to normal cells. Mechanistic studies revealed that Nano-S triggered inactivation of the MEK-ERK pathway followed by inhibition of the proliferation of the A375 and MCF-7 cells. In addition, lower Nano-S concentrations and shorter exposure stimulated the expression of a copper transporter as compensation, which further increased the cellular uptake and anticancer activities of cisplatin. Collectively, our results highlight the potential of Nano-S as an anticancer agent or adjuvant through its detention of copper.


Cell Proliferation/drug effects , Copper/metabolism , Nanoparticles/administration & dosage , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Sulfur/administration & dosage , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cisplatin/administration & dosage , Dose-Response Relationship, Drug , Drug Synergism , Humans , MCF-7 Cells , Neoplasms, Experimental/pathology , Treatment Outcome
4.
Chem Pharm Bull (Tokyo) ; 62(10): 994-9, 2014.
Article En | MEDLINE | ID: mdl-25273058

Selenadiazole derivatives are synthetic organoselenium compounds with improved anticancer activity and greater selectivity than inorganic selenium. In this study, 4-(benzo[c][1,2,5]selenadiazol-6-yl)-benzene-1,2-diamine (BSBD) was shown to induce time- and dose-dependent apoptosis in SWO-38 human glioma cells by accumulation of a sub-G1 cell population, DNA fragmentation, nuclear condensation, caspase activation and poly(ADP-ribose) polymerase (PARP) cleavage. Further mechanistic investigation showed that BSBD treatment induced dephosphorylation of AKT and DNA damage-mediated activation of p53, leading to extensive apoptosis through the mitochondrial pathway. Our findings suggest that BSBD represents a potential human glioma therapeutic.


Antineoplastic Agents/chemistry , Diamines/chemistry , Organoselenium Compounds/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , DNA Fragmentation/drug effects , Diamines/toxicity , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Glioma/metabolism , Glioma/pathology , Humans , Organoselenium Compounds/toxicity , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Tumor Suppressor Protein p53/metabolism
5.
Oncotarget ; 5(17): 7431-45, 2014 Sep 15.
Article En | MEDLINE | ID: mdl-25277183

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), as one of the most promising targeted drug for new cancer therapeutics, is limited in clinical application by the evolution of resistance in many cancer cell lines, especially in malignant melanoma. Thus, it is urgently needed to identify chemosensitizers to enhance the apoptotic inducing efficacy of TRAIL and overcome resistance of malignant melanoma cells. Herein, we reported that 3,3'-diselenodipropionic acid (DSeA), a Selenocysteine derivative, could synergistically enhance the growth inhibitory effect of TRAIL on A375 melanoma cells though induction of ROS-dependent apoptosis with involvement of PTEN-mediated Akt inactivation and DNA damage-mediated p53 phosphorylation, which subsequently activated mitochondrial and death receptor apoptotic pathways. Moreover, silencing of p53 down-regulated the expression levels of p53-inducible genes, and effectively blocked the cell apoptosis. Suppression of PI3K significantly increased the apoptotic cell death. In contrast, antioxidants effectively reversed the cell apoptosis through regulation of Akt and p53 signaling pathways. Taken together, the combination of DSeA and TRAIL could be a novel strategy to overcome TRAIL resistance in malignant melanoma, and DSeA may be candidates for further evaluation as a chemosensitizer in clinical trails.


Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Melanoma/metabolism , Propionates/pharmacology , Selenium Compounds/pharmacology , Skin Neoplasms/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Drug Synergism , Humans , In Situ Nick-End Labeling , RNA Interference , Reactive Oxygen Species/metabolism , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , TNF-Related Apoptosis-Inducing Ligand/pharmacology
6.
Mol Pharm ; 11(4): 1282-93, 2014 Apr 07.
Article En | MEDLINE | ID: mdl-24555485

Cisplatin-based therapy is one of the most important chemotherapy treatments for cancers. However, its efficacy is greatly limited by drug resistance and undesirable side effects. Therefore, it is of great importance to develop chemosensitizing agents to cisplatin. In the present study, we demonstrated the strategy to use methylseleninic acid (MeSe) as a synergistic agent of cisplatin and elucidated their action mechanisms. The combination of MeSe and cisplatin exhibited synergistic anticancer efficacy and achieved greater selectivity between cancer cell and normal cell. By inducing intracellular oxidative stress, MeSe potentiated cisplatin-induced DNA damage and led to enhanced p53 phosphorylation, followed by increased activation of both mitochondrial and death receptor pathway. Down-regulation of phosphorylated AKT and ERK also played important roles in the synergistic effects of MeSe and cisplatin. Our results suggested that the strategy to apply MeSe as a synergistic agent to cisplatin could be a highly efficient way to achieve anticancer synergism by targeting the intracellular redox system. MeSe might be a candidate for clinical application as a chemosensitizer to cisplatin-based therapy for cancer treatments, especially for hepatocellular carcinoma.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , Organoselenium Compounds/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/physiology , Cell Line, Tumor , Drug Synergism , Humans , Phosphorylation , Signal Transduction
7.
Int J Nanomedicine ; 7: 3939-49, 2012.
Article En | MEDLINE | ID: mdl-22915845

Gray selenium (Se) is one of the most widely used Se sources with very limited biocompatibility and bioactivity. In the present study, a simple method for the preparation of ultrasmall selenium nanoparticles (SeNPs) through direct nanolization of gray selenium by polyethylene glycol (PEG) was demonstrated. Monodisperse and homogeneous PEG-SeNPs with ultrasmall diameters were successfully prepared under optimized conditions. The products were characterized using various microscopic and spectroscopic methods, and the results suggest that the amphoteric properties of PEG and the coordination between oxygen and selenium atoms contributed to the formation of ultrasmall nanoparticles. PEG-SeNPs exhibited stronger growth inhibition on drug-resistant hepatocellular carcinoma (R-HepG2) cells than on normal HepG2 cells. Dose-dependent apoptosis was induced by PEG-SeNPs in R-HepG2 cells, as evidenced by an increase in the sub-G1 cell population. Further investigation on the underlying molecular mechanisms revealed that depletion of mitochondrial membrane potential and generation of superoxide anions contributed to PEG-SeNPs-induced apoptotic cell death in R-HepG2 cells. Our results suggest that PEG-SeNPs may be a candidate for further evaluation as a chemotherapeutic agent for drug-resistant liver cancer, and the strategy to use PEG200 as a surface decorator could be a highly efficient way to enhance the anticancer efficacy of nanomaterials.


Antineoplastic Agents/pharmacology , Membrane Potential, Mitochondrial/drug effects , Nanoparticles/chemistry , Polyethylene Glycols/pharmacology , Selenium/pharmacology , Analysis of Variance , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Carcinoma, Hepatocellular , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Hep G2 Cells , Humans , Liver Neoplasms , Nanotechnology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Selenium/chemistry , Selenium/pharmacokinetics , Superoxides/metabolism
8.
Colloids Surf B Biointerfaces ; 83(1): 183-7, 2011 Mar.
Article En | MEDLINE | ID: mdl-21145219

A simple method for fabrication of sialic acid surface-decorated selenium nanoparticles (SA-Se-NPs) with enhanced cancer-targeting and cell-penetrating abilities has been demonstrated in the present study. Monodisperse and homogeneous spherical SA-Se-NPs with striking stability were prepared under the optimized conditions. SA surface decoration significantly increased the cellular uptake and cytotoxicity of Se-NPs in HeLa human cervical carcinoma cells. Treatments of SA-Se-NPs induced dose-dependent apoptosis in HeLa cells, as evidenced by increase in sub-G1 cell populations, nuclear condensation and formation of apoptotic bodies. Further investigation on molecular mechanisms reveals that SA-Se-NPs triggered cancer cell apoptosis through activation of caspase-3 and subsequent cleavage of PARP.


Apoptosis/drug effects , N-Acetylneuraminic Acid/chemistry , Nanoparticles/chemistry , Selenium/metabolism , Selenium/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Drug Screening Assays, Antitumor , Flow Cytometry , HeLa Cells , Humans , Nanoparticles/ultrastructure , Surface Properties/drug effects
9.
Guang Pu Xue Yu Guang Pu Fen Xi ; 30(9): 2417-23, 2010 Sep.
Article Zh | MEDLINE | ID: mdl-21105409

The antioxidant activities of Dangcong tea aqueous extracts against DPPH and ABTS free radicals were evaluated using spectrometric methods. The results show that Dangcong tea aqueous extracts could effectively and rapidly inhibited the formation of ABTS and DPPH free radicals in a dose- and time-dependent manner, indicating the potent antioxidant activities of Dangcong tea aqueous extracts under both hydrophilic and hydrophobic conditions. In the optimized systems, the IC50 values of Baiye (I, II) and Fenghuang (III, IV) Dangcong teas against ABTS free radical were 26.9, 25.5, 28.0 and 31.7 microg x mL(-1), respectively, which were significantly lower than that of Trolox, the positive control (85.2 microg x mL(-1)), indicating the higher antioxidant activities of Dangcong teas. For DPPH free radical, the IC50 values for the Dangcong teas (I-IV) were 49.8, 41.6, 47.3 and 64.5 microg x mL(-1), respectively. Taken together, our results suggest the potential applications of Dangcong tea as functional food.


Free Radical Scavengers/chemistry , Plant Extracts/chemistry , Tea/chemistry , Benzothiazoles , Biphenyl Compounds , Free Radicals , Oxidation-Reduction , Picrates , Sulfonic Acids
...