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










Publication year range
1.
CNS Neurosci Ther ; 30(2): e14592, 2024 02.
Article in English | MEDLINE | ID: mdl-38385622

ABSTRACT

AIMS: Disturbances in the circadian rhythm are positively correlated with the processes of aging and related neurodegenerative diseases, which are also associated with brain iron accumulation. However, the role of brain iron in regulating the biological rhythm is poorly understood. In this study, we investigated the impact of brain iron levels on the spontaneous locomotor activity of mice with altered brain iron levels and further explored the potential mechanisms governing these effects in vitro. RESULTS: Our results revealed that conditional knockout of ferroportin 1 (Fpn1) in cerebral microvascular endothelial cells led to brain iron deficiency, subsequently resulting in enhanced locomotor activity and increased expression of clock genes, including the circadian locomotor output cycles kaput protein (Clock) and brain and muscle ARNT-like 1 (Bmal1). Concomitantly, the levels of period circadian regulator 1 (PER1), which functions as a transcriptional repressor in regulating biological rhythm, were decreased. Conversely, the elevated brain iron levels in APP/PS1 mice inhibited autonomous rhythmic activity. Additionally, our findings demonstrate a significant decrease in serum melatonin levels in Fpn1cdh5 -CKO mice compared with the Fpn1flox/flox group. In contrast, APP/PS1 mice with brain iron deposition exhibited higher serum melatonin levels than the WT group. Furthermore, in the human glioma cell line, U251, we observed reduced PER1 expression upon iron limitation by deferoxamine (DFO; iron chelator) or endogenous overexpression of FPN1. When U251 cells were made iron-replete by supplementation with ferric ammonium citrate (FAC) or increased iron import through transferrin receptor 1 (TfR1) overexpression, PER1 protein levels were increased. Additionally, we obtained similar results to U251 cells in mouse cerebellar astrocytes (MA-c), where we collected cells at different time points to investigate the rhythmic expression of core clock genes and the impact of DFO or FAC treatment on PER1 protein levels. CONCLUSION: These findings collectively suggest that altered iron levels influence the circadian rhythm by regulating PER1 expression and thereby modulating the molecular circadian clock. In conclusion, our study identifies the regulation of brain iron levels as a potential new target for treating age-related disruptions in the circadian rhythm.


Subject(s)
Iron , Melatonin , Mice , Humans , Animals , Iron/metabolism , Endothelial Cells/metabolism , Brain/metabolism , Circadian Rhythm/genetics , Period Circadian Proteins/genetics
2.
Cell Death Dis ; 15(1): 49, 2024 01 13.
Article in English | MEDLINE | ID: mdl-38218852

ABSTRACT

Transmembrane serine protease 6 (Tmprss6) has been correlated with the occurrence and progression of tumors, but any specific molecular mechanism linking the enzyme to oncogenesis has remained elusive thus far. In the present study, we found that Tmprss6 markedly inhibited mouse neuroblastoma N2a (neuro-2a) cell proliferation and tumor growth in nude mice. Tmprss6 inhibits Smad1/5/8 phosphorylation by cleaving the bone morphogenetic protein (BMP) co-receptor, hemojuvelin (HJV). Ordinarily, phosphorylated Smad1/5/8 binds to Smad4 for nuclear translocation, which stimulates the expression of hepcidin, ultimately decreasing the export of iron through ferroportin 1 (FPN1). The decrease in cellular iron levels in neuro-2a cells with elevated Tmprss6 expression limited the availability of the metal forribo nucleotide reductase activity, thereby arresting the cell cycle prior to S phase. Interestingly, Smad4 promoted nuclear translocation of activating transcription factor 3 (ATF3) to activate the p38 mitogen-activated protein kinases signaling pathway by binding to ATF3, inducing apoptosis of neuro-2a cells and inhibiting tumor growth. Disruption of ATF3 expression significantly decreased apoptosis in Tmprss6 overexpressed neuro-2a cells. Our study describes a mechanism whereby Tmprss6 regulates the cell cycle and apoptosis. Thus, we propose Tmprss6 as a candidate target for inhibiting neuronal tumor growth.


Subject(s)
Hepcidins , Neoplasms , Animals , Mice , Bone Morphogenetic Proteins/metabolism , Iron/metabolism , Mice, Nude
3.
Int J Neurosci ; : 1-14, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37937941

ABSTRACT

Nerve injury can be caused by a variety of factors. It often takes a long time to repair a nerve injury and severe nerve injury is even difficult to heal. Therefore, increasing attention has focused on nerve injury and repair. Long non-coding RNA (lncRNA) is a newly discovered non-coding RNA with a wide range of biological activities. Numerous studies have shown that a variety of lncRNAs undergo changes in expression after nerve injury, indicating that lncRNAs may be involved in various biological processes of nerve repair and regeneration. Herein, we summarize the biological roles of lncRNAs in neurons, glial cells and other cells during nerve injury and regeneration, which will help lncRNAs to be better applied in nerve injury and regeneration in the future.

4.
Redox Biol ; 57: 102475, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36179435

ABSTRACT

Oxidative stress and deficient bioenergetics are key players in the pathological process of cerebral ischemia reperfusion injury (I/R). As a mitochondrial iron storage protein, mitochondrial ferritin (FtMt) plays a pivotal role in protecting neuronal cells from oxidative damage under stress conditions. However, the effects of FtMt in mitochondrial function and activation of apoptosis under cerebral I/R are barely understood. In the present study, we found that FtMt deficiency exacerbates neuronal apoptosis via classical mitochondria-depedent pathway and the endoplasmic reticulum (ER) stress pathway in brains exposed to I/R. Conversely, FtMt overexpression significantly inhibited oxygen and glucose deprivation and reperfusion (OGD/R)-induced apoptosis and the activation of ER stress response. Meanwhile, FtMt overexpression rescued OGD/R-induced mitochondrial iron overload, mitochondrial dysfunction, the generation of reactive oxygen species (ROS) and increased neuronal GSH content. Using the Seahorse and O2K cellular respiration analyser, we demonstrated that FtMt remarkably improved the ATP content and the spare respiratory capacity under I/R conditions. Importantly, we found that glucose consumption was augmented in FtMt overexpressing cells after OGD/R insult; overexpression of FtMt facilitated the activation of glucose 6-phosphate dehydrogenase and the production of NADPH in cells after OGD/R, indicating that the pentose-phosphate pathway is enhanced in FtMt overexpressing cells, thus strengthening the antioxidant capacity of neuronal cells. In summary, our results reveal that FtMt protects against I/R-induced apoptosis through enhancing mitochondrial bioenergetics and regulating glucose metabolism via the pentose-phosphate pathway, thus preventing ROS overproduction, and preserving energy metabolism.

5.
Antioxidants (Basel) ; 11(7)2022 Jun 26.
Article in English | MEDLINE | ID: mdl-35883748

ABSTRACT

Blood-brain barrier (BBB) breakdown, a characteristic feature of ischemic stroke, contributes to poor patient outcomes. Brain microvascular endothelial cells (BMVECs) are a key component of the BBB and dysfunction or death of these cells following cerebral ischemia reperfusion (I/R) injury can disrupt the BBB, leading to leukocyte infiltration, brain edema and intracerebral hemorrhage. We previously demonstrated that mitochondrial ferritin (FtMt) can alleviate I/R-induced neuronal ferroptosis by inhibiting inflammation-regulated iron deposition. However, whether FtMt is involved in BBB disruption during cerebral I/R is still unknown. In the present study, we found that FtMt expression in BMVECs is upregulated after I/R and overexpression of FtMt attenuates I/R-induced BBB disruption. Mechanistically, we found that FtMt prevents tight junction loss and apoptosis by inhibiting iron dysregulation and reactive oxygen species (ROS) accumulation in I/R-treated BMVECs. Chelating excess iron with deferoxamine alleviates apoptosis in the brain endothelial cell line bEnd.3 under oxygen glucose deprivation followed by reoxygenation (OGD/R) insult. In summary, our data identify a previously unexplored effect for FtMt in the BBB and provide evidence that iron-mediated oxidative stress in BMVECs is an early cause of BMVECs damage and BBB breakdown in ischemic stroke.

6.
Cell Signal ; 96: 110361, 2022 08.
Article in English | MEDLINE | ID: mdl-35643226

ABSTRACT

By improving the previous method of CUT&RUN, we developed D-CUT&RUN (DSP fixed CUT&RUN) for under-expressed transcription factor. High-quality data could be obtained for low expressed transcription factors using chemical crosslinkers (DSP) and reducing agent (DTT). We applied our D-CUT&RUN to detection of Bcl11b and Mycn binding sites in mammary epithelial progenitor cells. Pathway enrichment analysis results of Bcl11b target genes showed that Bcl11b was a regulatory factor involved in breast cancer and it could negatively regulate Wnt signaling pathway. Furthermore, the role of Bcl11b in breast cancer was mediated by catabolic process and stress-related pathway. Our research suggested that D-CUT&RUN could be used for low abundance transcription factor binding sites detection and Bcl11b could be a target for breast cancer treatment in the future.


Subject(s)
Breast Neoplasms , Transcription Factors , Breast Neoplasms/genetics , Female , Humans , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway
7.
Bioact Mater ; 18: 526-538, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35415298

ABSTRACT

Despite the progress made to improve therapeutic outcomes for acute myeloid leukemia (AML), many unmet clinical needs remain to be resolved. Unlike existing anti-AML strategies, here we developed a biomimetic nanocomposite to efficiently eliminate the leukemia cells in the bone marrow and prevent the homing of AML. To fulfill our design, the ultra-small nanozyme was conjugated onto the surface of an oxygen-carrying nanoparticle, which was further coated with bone marrow stromal cell membrane. After entering the blood, this biomimetic nanocomposite got actively internalized by the leukemia cells in the blood and released the loaded chemotherapeutics and nanozyme inside the leukemia cells to achieve a synergistic antitumor efficacy. Meanwhile, the adhesive properties of the stromal cell membrane enabled the nanocomposite to home to the bone marrow, where the nanocomposite effectively killed the retained leukemia cells. More importantly, the biomimetic cell membrane also acted as a CXCR4 antagonism to block the CXCR4/CXCL12-mediated homing of leukemia cells to the bone marrow and infiltration to other organs like the liver and spleen. In conclusion, this proof-of-concept study demonstrated that our designed platform effectively kills leukemia cells while preventing their infiltration, thus providing a promising prospect for resolving the clinical challenges in current AML treatment.

8.
ACS Appl Mater Interfaces ; 13(36): 42382-42395, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34473471

ABSTRACT

Anthracycline-induced liver injury (AILI) is becoming an increasingly serious and potential clinical complication and is linked to reactive oxygen species (ROS) production and subsequent inflammatory response. Herein, we demonstrated that artificial Prussian blue nanozymes (PBZs) prevented daunorubicin-induced liver injury, a prototype of AILI, by attenuating ROS production and regulating inflammation. PBZs exhibited multienzyme activity and could scavenge ROS and free radicals. At the cellular level, PBZs could effectively eliminate ROS, suppress hepatocyte apoptosis, reduce deoxyribonucleic acid damage, and decrease the levels of inflammatory cytokines and chemokines. According to the results of the in vivo study, pretreatment with PBZs also resulted in a desirable protective effect against AILI, as indicated by both a decrease in biochemical indicator levels and hepatocyte necrosis. PBZs upregulated antioxidative genes by activating the Nrf2 pathway to reduce oxidative stress. Meanwhile, PBZs counteracted the inflammatory response based on the decreased expression levels of myeloperoxidase and F4/80 in the liver. Collectively, our findings indicate that PBZ-based nanotherapy is a novel strategy for protecting against AILI.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Daunorubicin/toxicity , Free Radical Scavengers/therapeutic use , Inflammation/drug therapy , Nanoparticles/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/toxicity , Apoptosis/drug effects , Catalysis , Chemical and Drug Induced Liver Injury/metabolism , Chemokines/metabolism , DNA/drug effects , DNA Damage/drug effects , Ferrocyanides/chemistry , Ferrocyanides/therapeutic use , Ferrocyanides/toxicity , Free Radical Scavengers/chemistry , Free Radical Scavengers/toxicity , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/toxicity , Oxidative Stress/drug effects , Povidone/chemistry , Povidone/toxicity , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
9.
J Mater Chem B ; 9(26): 5245-5254, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34095945

ABSTRACT

Multifunctional drug delivery systems combining two or more therapies have a wide-range of potential for high efficacy tumor treatment. Herein, we designed a novel hollow mesoporous Prussian blue nanoparticles (HMPBs)-based platform for targeted and synergetic chemo-photothermal treatment of acute myeloid leukemia (AML). The HMPBs were first loaded with the anticancer drugs daunorubicin (DNR) and cytarabine (AraC), and were subsequently coated with polyethylenimine (PEI) through electrostatic adsorption. Then, zwitterionic sulfobetaine (ZS) and CXCR4 antagonist peptide E5 were modified onto the surface of the nanoparticles via covalent bonding to fabricate a nanoplatform (denoted as HMPBs(DNR + AraC)@PEI-ZS-E5). The nanoplatform showed excellent photothermal effects, superior photothermal stability, reduced nonspecific protein adsorption, efficient targeting capability, a constant hydrodynamic diameter and good biocompatibility. Additionally, a laser-responsive drug release pattern was observed. In vitro results indicated that the nanoplatform could achieve active targeting and remarkable chemo-photothermal synergetic therapeutic effects, showcasing its great potential in AML treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cytarabine/pharmacology , Daunorubicin/pharmacology , Ferrocyanides/chemistry , Leukemia, Myeloid, Acute/drug therapy , Nanoparticles/chemistry , Photothermal Therapy , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytarabine/chemistry , Daunorubicin/chemistry , Drug Delivery Systems , Drug Screening Assays, Antitumor , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/pathology , Particle Size , Polyethyleneimine/chemistry , Porosity , Surface Properties
10.
Analyst ; 145(6): 2367-2377, 2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32031182

ABSTRACT

Salmonella spp. are zoonotic pathogens of substantial public health concern. To enable detection in the field or under instrument-free conditions, we developed a rapid and robust lateral flow fluorescent immunoassay based on strand exchange amplification (SEA-LFIA) for the quantitative detection of Salmonella spp. As far as we know, this work is the first report regarding the use of Bst DNA polymerase-assisted SEA for fluorescence sensing to detect Salmonella spp. The SEA method was further confirmed by enzymatic digestion and Sanger dideoxy sequencing. The specificity of SEA-LFIA assay was verified by 89 Salmonella strains (18 Salmonella reference strains and 71 clinical isolates) and 15 non-Salmonella reference strains (different genera). The sensitivity of SEA-LFIA assay was 6 × 100 CFU mL-1 of Salmonella pure culture or 3 × 104 CFU 25 g-1 of artificially spiked raw chicken meat. Using this assay, it was found that 37 (16%) of the 236 samples collected were positive, which was consistent with the results of conventional PCR. The cutoff value is 15 and SEA-LFIA assay only takes ∼30 min without high equipment and reagent cost. In addition, the proposed strategy can be easily extended by redesigning the corresponding amplification primers to detect target analytes. In conclusion, the optimized SEA-LFIA assay is an efficient and specific method for the detection of Salmonella spp., and can potentially serve as a new on-site diagnostic tool in life sciences.


Subject(s)
Fluoroimmunoassay/methods , Poultry/microbiology , Salmonella Infections/microbiology , Salmonella/isolation & purification , Animals , Chickens/microbiology , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Equipment Design , Fluorescent Antibody Technique/economics , Fluorescent Antibody Technique/methods , Fluoroimmunoassay/economics , Food Analysis/economics , Food Analysis/methods , Food Contamination/analysis , Humans , Nucleic Acid Amplification Techniques/economics , Nucleic Acid Amplification Techniques/methods , Salmonella/genetics , Time Factors
11.
Transl Res ; 196: 31-41, 2018 06.
Article in English | MEDLINE | ID: mdl-29548626

ABSTRACT

Drug-induced liver injury shares a common feature of mitochondrial dysfunction. Mitochondrial therapy (mitotherapy), which replaces malfunctional mitochondria with functional exogenous mitochondria, may be a fundamental approach for treating drug-mediated hepatotoxicity. Here, we suggested that mitochondria isolated from human hepatoma cell could be used to treat acetaminophen (APAP)-induced liver injury in mice. When the mitochondria were added into the cell media, they could enter primarily cultured mouse hepatocyte. When the mitochondria were intravenously injected into mice, they distribute in several tissues, including liver. In the model mice of APAP-induced liver injury, mitochondria treatment increased hepatocyte energy supply, reduced oxidation stress, and consequently ameliorated tissue injury. The study suggests that exogenous mitochondria could be an effective therapeutic strategy in treating APAP-induced liver injury.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Mitochondria/physiology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Chemical and Drug Induced Liver Injury/metabolism , Disease Models, Animal , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Oxidative Stress
12.
Yao Xue Xue Bao ; 52(3): 390-6, 2017 Mar.
Article in Chinese | MEDLINE | ID: mdl-29979701

ABSTRACT

This study was conducted to test the effects of schizandrin B (Sch B) on clozapine (CLZ) induced chronic liver injury in mice and the mechanism of action, and this may provide a new approach for clinical prevention of CLZ-induced side effects. The CLZ was given to mice for three weeks alone or co-administration with Sch B. The changes of alanine aminotransferase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP) and antioxidation indexes superoxide dismutase (SOD), malonic dialdehyde (MDA), glutathione (GSH) and liver histological evaluation were determined. Expression of Nrf2 was assayed in hepatic cells by immunohistochemical staining and Western blotting. The changes of relative gene expression of NAD(P)H: quinone oxidoreductase l (NQO1) and heme oxygenase 1 (HO-1) were assayed by real-time Q-PCR. The results showed that pretreatment with a lower dosage of Sch B (25, 50 mg·kg−1) prevented CLZ-induced liver injury as indicated by the reduced levels of ALT, AST and ALP, and the preserved activities of SOD, GSH and inhibiting MDA. It was shown that Sch B could up-regulate Nrf2 expression leading to nuclear accumulation of Nrf2 to induce oxidative response genes such as NQO1 and HO-1. These results suggest that Sch B could protect against liver injury induced by CLZ via the activation of the Nrf2/ARE signal pathway in a dose-dependent manner.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Clozapine/adverse effects , Lignans/pharmacology , Polycyclic Compounds/pharmacology , Alanine Transaminase/metabolism , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/metabolism , Cyclooctanes/pharmacology , Glutathione/metabolism , Heme Oxygenase-1/metabolism , Liver/drug effects , Malondialdehyde/metabolism , Membrane Proteins/metabolism , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Protective Agents/pharmacology , Signal Transduction , Superoxide Dismutase/metabolism
13.
Oncogene ; 35(38): 5056-5069, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27041584

ABSTRACT

Melanoma is one of the most lethal forms of skin cancer because of its early metastatic spread. The variant form of CD44 (CD44v), a cell surface glycoprotein, is highly expressed on metastatic melanoma. The mechanisms of regulation of CD44 alternative splicing in melanoma and its pathogenic contributions are so far poorly understood. Here, we investigated the expression level of CD44 in a large set of melanocytic lesions at different stages. We found that the expression of CD44v8-10 and a splicing factor, U2AF2, is significantly increased during melanoma progression, whereas CD82/KAI1, a tetraspanin family of tumor suppressor, is reduced in metastatic melanoma. CD44v8-10 and U2AF2 expression levels, which are negatively correlated with CD82 levels, are markedly elevated in primary melanoma compared with dysplastic nevi and further increased in metastatic melanoma. We also showed that patients with higher CD44v8-10 and U2AF2 expression levels tended to have shorter survival. By using both in vivo and in vitro assays, we demonstrated that CD82 inhibits the production of CD44v8-10 on melanoma. Mechanistically, U2AF2 is a downstream target of CD82 and in malignant melanoma facilitates CD44v8-10 alternative splicing. U2AF2-mediated CD44 isoform switch is required for melanoma migration in vitro and lung and liver metastasis in vivo. Notably, overexpression of CD82 suppresses U2AF2 activity by inducing U2AF2 ubiquitination. In addition, our data suggested that enhancement of melanoma migration by U2AF2-dependent CD44v8-10 splicing is mediated by Src/focal adhesion kinase/RhoA activation and formation of stress fibers, as well as CD44-E-selectin binding reinforcement. These findings uncovered a hitherto unappreciated function of CD82 in severing the linkage between U2AF2-mediated CD44 alternative splicing and cancer aggressiveness, with potential prognostic and therapeutic implications in melanoma.


Subject(s)
Hyaluronan Receptors/genetics , Kangai-1 Protein/genetics , Melanoma/genetics , Splicing Factor U2AF/genetics , Alternative Splicing/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Melanocytes/pathology , Melanoma/pathology , Mice , Neoplasm Metastasis , Phosphorylation , Prognosis , Protein Isoforms/genetics , Proteolysis , Ubiquitination/genetics , Xenograft Model Antitumor Assays
14.
J Biol Chem ; 291(5): 2087-106, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26504080

ABSTRACT

Tumor invasiveness depends on the ability of tumor cells to breach endothelial barriers. In this study, we investigated the mechanism by which the adhesion of melanoma cells to endothelium regulates adherens junction integrity and modulates tumor transendothelial migration (TEM) by initiating thrombin generation. We found that the B-Raf(V600E) mutation in metastatic melanoma cells up-regulated tissue factor (TF) expression on cell membranes and promoted thrombin production. Co-culture of endothelial monolayers with metastatic melanoma cells mediated the opening of inter-endothelial spaces near melanoma cell contact sites in the presence of platelet-free plasma (PFP). By using small interfering RNA (siRNA), we demonstrated that B-Raf(V600E) and TF silencing attenuated the focal disassembly of adherens junction induced by tumor contact. Vascular endothelial-cadherin (VE-cadherin) disassembly was dependent on phosphorylation of p120-catenin on Ser-879 and VE-cadherin on Tyr-658, Tyr-685, and Tyr-731, which can be prevented by treatment with the thrombin inhibitor, hirudin, or by silencing the thrombin receptor, protease-activated receptor-1, in endothelial cells. We also provided strong evidence that tumor-derived thrombin enhanced melanoma TEM by inducing ubiquitination-coupled VE-cadherin internalization, focal adhesion formation, and actin assembly in endothelium. Confocal microscopic analysis of tumor TEM revealed that junctions transiently opened and resealed as tumor cells accomplished TEM. In addition, in the presence of PFP, tumor cells preferentially transmigrated via paracellular routes. PFP supported melanoma transmigration under shear conditions via a B-Raf(V600E)-thrombin-dependent mechanism. We concluded that the activation of thrombin generation by cancer cells in plasma is an important process regulating melanoma extravasation by disrupting endothelial junction integrity.


Subject(s)
Endothelium, Vascular/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/metabolism , Thrombin/metabolism , Thromboplastin/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Electric Impedance , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Mutation , Neoplasm Metastasis , Permeability , Phosphorylation , Plasma/metabolism , RNA, Small Interfering/metabolism , Ubiquitination
15.
Chem Res Toxicol ; 28(4): 765-74, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25742418

ABSTRACT

Tetrachlorobenzoquinone (TCBQ), a metabolite of industrial herbicide pentachlorophenol, showed hepatotoxicity and genotoxicity through reactive oxygen species (ROS) mechanism in vivo and in vitro models. Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a cellular sensor of oxidative or electrophilic stress, which controls the expression of detoxifying enzymes and antioxidant proteins. Using the human hepatoma HepG2 cell line as an in vitro model, we demonstrated a significant induction of Nrf2 but not its negative regulator Kelch-like ECH-associated protein 1 (Keap1), following exposure to TCBQ. Also, our results clearly demonstrated the translocation of cytosolic Nrf2 into the nucleus. After translocation, Nrf2 subsequently binds to the antioxidant response element (ARE), up-regulated heme oxygenase-1 (HO-1), and NADH quinone oxidoreductase subunit 1 (NQO1), which may be considered as an antioxidative response to TCBQ-intoxication. The luciferase reporter assay confirmed the formation of the Nrf2-ARE complex. Furthermore, mechanism studies proposed that TCBQ promoted the formation of the Keap1 cross-linking dimer, a ubiquitination switch from Nrf2 to Keap1 but not the dissociation of the Keap1-Cullin3 (Cul3) complex.


Subject(s)
Benzoquinones/toxicity , Cullin Proteins/metabolism , Hydrocarbons, Chlorinated/toxicity , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Ubiquitin/metabolism , Dimerization , Hep G2 Cells , Humans , Kelch-Like ECH-Associated Protein 1 , Protein Processing, Post-Translational , Protein Transport , RNA Processing, Post-Transcriptional , Ubiquitination
16.
Toxicol Lett ; 234(3): 151-61, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25725129

ABSTRACT

Diphenyl difluoroketone (EF24), a curcumin analog, exhibits potent anti-tumor activities by arresting cell cycle and inducing apoptosis. However, the efficacy and modes of action of EF24 on melanoma metastasis remain elusive. In this study, we found that at non-cytotoxic concentrations, EF24 suppressed cell motility and epithelial-to-mesenchymal Transition (EMT) of melanoma cell lines, Lu1205 and A375. EF24 also suppressed HMGA2 expression at mRNA and protein levels. miR-33b directly bound to HMGA2 3' untranslated region (3'-UTR) to suppress its expression as measured by dual-luciferase assay. EF24 increased expression of E-cadherin and decreased STAT3 phosphorylation and expression of the mesenchymal markers, vimentin and N-cadherin. miR-33b inhibition or HMGA2 overexpression reverted EF24-mediated suppression of EMT phenotypes. In addition, EF24 modulated the HMGA2-dependent actin stress fiber formation, focal adhesion assembly and FAK, Src and RhoA activation by targeting miR-33b. Thus, the results suggest that EF24 suppresses melanoma metastasis via upregulating miR-33b and concomitantly reducing HMGA2 expression. The observed activities of EF24 support its further evaluation as an anti-metastatic agent in melanoma therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzylidene Compounds/pharmacology , Cell Movement/physiology , Curcumin/analogs & derivatives , Epithelial-Mesenchymal Transition/drug effects , HMGA2 Protein/drug effects , Melanoma/metabolism , MicroRNAs/drug effects , Piperidones/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/physiology , Fluorescent Antibody Technique , Humans , Melanoma/drug therapy , Melanoma/physiopathology , MicroRNAs/physiology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
17.
Am J Physiol Heart Circ Physiol ; 308(10): H1205-14, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25770237

ABSTRACT

Environmental hazardous material polychlorinated biphenyl (PCB) exposure is associated with vascular endothelial dysfunction, which may increase the risk of cardiovascular diseases and cancer metastasis. Our previous studies illustrated the cytotoxic, antiproliferative, and genotoxic effects of a synthetic, quinone-type, highly reactive metabolite of PCB, 2,3,5-trichloro-6-phenyl-[1,4]benzoquinone (PCB29-pQ). Here, we used it as the model compound to investigate its effects on vascular endothelial integrity and permeability. We demonstrated that noncytotoxic doses of PCB29-pQ induced vascular endothelial (VE)-cadherin junction disassembly by increasing the phosphorylation of VE-cadherin at Y658. We also found that focal adhesion assembly was required for PCB29-pQ-induced junction breakdown. Focal adhesion site-associated actin stress fibers may serve as holding points for cytoskeletal tension to regulate the cellular contractility. PCB29-pQ exposure promoted the association of actin stress fibers with paxillin-containing focal adhesion sites and enlarged the size/number of focal adhesions. In addition, PCB29-pQ treatment induced phosphorylation of paxillin at Y118. By using pharmacological inhibition, we further demonstrated that p38 activation was necessary for paxillin phosphorylation, whereas extracellular signal-regulated kinases-1/2 activation regulated VE-cadherin phosphorylation. In conclusion, these results indicated that PCB29-pQ stimulates endothelial hyperpermeability by mediating VE-cadherin disassembly, junction breakdown, and focal adhesion formation. Intervention strategies targeting focal adhesion and MAPK signaling could be used as therapeutic approaches for preventing adverse cardiovascular health effects induced by environmental toxicants such as PCBs.


Subject(s)
Antigens, CD/metabolism , Benzoquinones/pharmacology , Cadherins/metabolism , Focal Adhesions/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , MAP Kinase Signaling System , Polychlorinated Biphenyls/pharmacology , Antigens, CD/genetics , Cadherins/genetics , Capillary Permeability , Focal Adhesions/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Paxillin/metabolism
19.
Biochem Biophys Res Commun ; 459(1): 1-9, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25475728

ABSTRACT

Melanoma cell migration across vascular endothelial cells is an essential step of tumor metastasis. Here, we provide evidence that RacGAP1, a cytokinesis-related Rho GTPase-activating protein, contributed to this process. Depletion of RacGAP1 with RacGAP1-targeting siRNA or overexpression of RacGAP1 mutant (T249A) attenuated melanoma cell transendothelial migration and concomitant changes of adherens junctions. In addition, RacGAP1 promoted the activations of RhoA, FAK, paxillin and triggered focal adhesion formation and cytoskeletal rearrangement. By overexpressing FAK-related non-kinase (FRNK) in endothelium, we showed that RacGAP1 mediated endothelial barrier function loss and melanoma transmigration in a focal adhesion-dependent manner. These results suggest that endothelial RacGAP1 may play critical roles in pathogenic processes of cancer by regulating endothelial permeability.


Subject(s)
Adherens Junctions/metabolism , GTPase-Activating Proteins/metabolism , Melanoma/pathology , Transendothelial and Transepithelial Migration , Cell Line, Tumor , Cell Movement , Endothelial Cells/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/metabolism , GTPase-Activating Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Melanoma/metabolism , Mutation , Paxillin/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
20.
FEBS Lett ; 588(24): 4573-82, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25447529

ABSTRACT

Loss of endothelial adherens junctions is involved in tumor metastasis. Here, we demonstrate that, in the metastatic Lu1205 melanoma cells, expression of the CD44 variant CD44v8-v10 induced junction disassembly and vascular endothelial (VE)-cadherin phosphorylation at Y658 and Y731. Short interfering RNA (siRNA)-mediated CD44 knockdown or sialic acid cleavage reversed these effects. Moreover, microspheres coated with recombinant CD44v8-v10 promoted endothelial junction disruption. Overexpression of CD44v8-v10 but not of standard CD44 (CD44s) promoted gap formation in the non-metastatic WM35 melanoma cells, whereas CD44 knockdown or neuraminidase treatment dramatically diminished melanoma transendothelial migration. Endothelial cells transfected with the phosphomimetic VE-cadherin mutant Y658E supported transmigration of CD44-silenced Lu1205 cells. Our findings imply that CD44 variant isoform (CD44v) but not CD44s regulates endothelial junction loss, promoting melanoma extravasation.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Endothelial Cells/pathology , Hyaluronan Receptors/metabolism , Melanoma/pathology , Adherens Junctions/drug effects , Cell Adhesion , Cell Line, Tumor , Cell Movement , Gene Knockdown Techniques , Humans , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/genetics , N-Acetylneuraminic Acid/metabolism , Neoplasm Metastasis , Phosphorylation , Protein Isoforms/chemistry , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...