Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 322
Filter
1.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(1): 60-66, 2024 Jan 20.
Article in Chinese | MEDLINE | ID: mdl-38322534

ABSTRACT

Objective: To explore the relationship between the expression of plectin and the migration of hepatocellular carcinoma (HCC) cells and to elucidate the molecular mechanisms by which plectin expression affects the migration of HCC cells. Methods: First of all, Western blot was performed to determine the expression of plectin in normal hepatocytes and HCC cells. Secondly, a plectin-downregulated HCC cell strain was established and the control group (shNC group) and shPLEC group were set up. Each group was divided into a vehicle control group (shNC+DMSO group or shPLEC+DMSO group) and a F-actin cytoskeleton polymerization inducer Jasplakinolide group (shNC+Jasp group or shPLEC+Jasp group). Western blot was performed to determine the expression of plectin and epithelial-mesenchymal transition (EMT)-related proteins, including N-cadherin, vimentin, and E-cadherin. HCC cell migration was evaluated by Transwell assay. KEGG (Kyoto Encyclopedia of Genes and Genomes) was used to analyze the signaling pathways related to plectin gene. The polymerization of F-actin was analyzed by immunofluorescence assay. Results: Compared with the normal hepatocytes, HCC cells showed high expression of plectin. Compared with those in the shNC group, the expression of plectin in the shPLEC group was decreased (P<0.05), the migration ability of HCC cells was weakened (P<0.05), and the EMT process was inhibited (with the expression of N-cadherin and vimentin being decreased and the expression of E-cadherin being increased) (P<0.05). KEGG analysis showed that the regulation of cytoskeletal F-actin was most closely associated with plectin and cytoskeletal F-actin depolymerized in the shPLEC group. After treatment with Jasplakinolide, an inducer of F-actin cytoskeleton polymerization, the migration ability of HCC cells in the shPLEC+Jasp group was enhanced compared with that of shPLEC+DMSO group (P<0.05) and the EMT process was restored (with the expression of N-cadherin and vimentin being increased and the expression of E-cadherin being decreased) (P<0.05). In addition, the polymerization of cytoskeletal F-actin in HCC cells was also restored. Conclusion: Plectin is highly expressed in HCC cells. Plectin promotes the migration and the EMT of HCC cells through inducing F-actin polymerization.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Plectin , Humans , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Actins/metabolism , Cadherins/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Dimethyl Sulfoxide , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Plectin/genetics , Plectin/metabolism , Polymerization , Vimentin/metabolism
2.
Arch Med Res ; 54(7): 102894, 2023 11.
Article in English | MEDLINE | ID: mdl-37806182

ABSTRACT

BACKGROUND: Numerous studies have confirmed that the leucine zipper tumor suppressor (LZTS) gene family plays a vital role in modulating transcription and cell cycle control, especially in colorectal cancer. This study aimed to evaluate the potential of leucine zipper tumor suppressor family member 3 (LZTS3) as a marker for COAD. METHODS: Bioinformatics, immunohistochemistry, and Western blotting were applied to assess the expression of LZTS3 in tissues. Gene overexpression or silencing was used to examine the biological roles of LZTS3 and validated using an in vivo nude mouse-human tumor model. RESULTS: The results obtained in this study indicate that LZTS3 is highly expressed in COAD. RTCA, Transwell, actin stain, and in vitro transfection experiments confirmed that LZTS3 expression inhibits tumor cell proliferation and cell migration. The results obtained in the nude mouse-human tumor model are consistent with those obtained in vitro. In particular, LZTS3 may exert biological effects by targeting the NOTCH signaling pathway. Furthermore, TAGLN was demonstrated to be a downstream target of LZTS3. CONCLUSION: This is the first study to demonstrate the important role of LZTS3 in the proliferation and migration of COAD and to shed light on the molecular mechanism underlying the tumor-suppressing role of LZTS3.


Subject(s)
Adenocarcinoma , Colorectal Neoplasms , Animals , Humans , Mice , Actin Cytoskeleton/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Mice, Nude , Tumor Suppressor Proteins/genetics
3.
Biol Direct ; 18(1): 41, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37501163

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe form of muscular dystrophy caused by mutations in the dystrophin gene. We characterized which isoforms of dystrophin were expressed by human induced pluripotent stem cell (hiPSC)-derived cardiac fibroblasts obtained from control and DMD patients. Distinct dystrophin isoforms were observed; however, highest molecular weight isoform was absent in DMD patients carrying exon deletions or mutations in the dystrophin gene. The loss of the full-length dystrophin isoform in hiPSC-derived cardiac fibroblasts from DMD patients resulted in deficient formation of actin microfilaments and a metabolic switch from mitochondrial oxidation to glycolysis. The DMD hiPSC-derived cardiac fibroblasts exhibited a dysregulated mitochondria network and reduced mitochondrial respiration, with enhanced compensatory glycolysis to sustain cellular ATP production. This metabolic remodeling was associated with an exacerbated myofibroblast phenotype and increased fibroblast activation in response to pro fibrotic challenges. As cardiac fibrosis is a critical pathological feature of the DMD heart, the myofibroblast phenotype induced by the absence of dystrophin may contribute to deterioration in cardiac function. Our study highlights the relationship between cytoskeletal dynamics, metabolism of the cell and myofibroblast differentiation and provides a new mechanism by which inactivation of dystrophin in non-cardiomyocyte cells may increase the severity of cardiopathy.


Subject(s)
Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Humans , Dystrophin/genetics , Dystrophin/metabolism , Myocytes, Cardiac/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Phenotype , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Fibroblasts/metabolism , Fibrosis , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
Med Oncol ; 40(3): 85, 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36692844

ABSTRACT

Medulloblastoma (MB) is children's most common primary malignant primitive neuro-ectodermal tumor. Group 3 MB showed a higher propensity to metastasis, which is molecularly characterized by c-MYC gene amplification. The activation of c-MYC promotes the remodeling of the F-actin cytoskeleton to enhance metastasis. The B7 homologue 6 (B7-H6) is associated with the manifold essential hallmarks of tumorigenesis. In this study, we will explore whether B7-H6 regulates the reorganization of F-actin by elevating the c-MYC expression to promote metastasis. The Daoy cell line was used to act as the cell model of medulloblastoma. Small interfering RNA and the plasmid were used to downregulate and upregulate the expression of B7-H6 in Daoy cells. Transwell assays with/without the matrigel matrix were used to detect migration and invasion of Daoy cells. Western blots were used to detect the expression of related proteins. Immunofluorescence staining was used to observe the impact of B7-H6 on the c-MYC /F-actin axis. B7-H6 improved migration and invasion in the Daoy cell line. B7-H6 enhanced the rearrangement of F-actin and activated the expression of MMP-9 and MMP-2. B7-H6 promoted the remodeling of F-actin by targeting c-MYC activation to reinforce migration and invasion. B7-H6 acts as a promoter of migration and invasion in medulloblastoma by activating the c-MYC /F-actin axis.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Child , Humans , Medulloblastoma/genetics , Medulloblastoma/metabolism , Medulloblastoma/pathology , Actins/genetics , Actins/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Cell Adhesion Molecules/metabolism , B7-1 Antigen/metabolism , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Cell Line, Tumor
5.
Sci Rep ; 12(1): 1846, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115632

ABSTRACT

Adolescent idiopathic scoliosis (AIS) is the most prevalent pediatric spinal deformity. We previously demonstrated elongated cilia and an altered molecular mechanosensory response in AIS osteoblasts. The purpose of this exploratory study was to characterize the mechanosensory defect occurring in AIS osteoblasts. We found that cilia length dynamics in response to flow significantly differ in AIS osteoblasts compared to control cells. In addition, strain-induced rearrangement of actin filaments was compromised resulting in a failure of AIS osteoblasts to position or elongate in function of the bidirectional-applied flow. Contrary to control osteoblasts, fluid flow had an inhibitory effect on AIS cell migration. Moreover, flow induced an increase in secreted VEGF-A and PGE2 in control but not AIS cells. Collectively our data demonstrated that in addition to the observed primary cilium defects, there are cytoskeletal abnormalities correlated to impaired mechanotransduction in AIS. Thus, we propose that the AIS etiology could be a result of generalized defects in cellular mechanotransduction given that an adolescent growing spine is under constant stimulation for growth and bone remodeling in response to applied mechanical forces. Recognition of an altered mechanotransduction as part of the AIS pathomechanism must be considered in the conception and development of more effective bracing treatments.


Subject(s)
Actin Cytoskeleton/metabolism , Cilia/metabolism , Mechanotransduction, Cellular , Osteoblasts/metabolism , Scoliosis/metabolism , Spine/metabolism , Actin Cytoskeleton/pathology , Adolescent , Braces , Case-Control Studies , Cell Movement , Cells, Cultured , Child , Cilia/pathology , Dinoprostone/metabolism , Female , Humans , Osteoblasts/pathology , Scoliosis/pathology , Scoliosis/therapy , Spine/pathology , Stress, Mechanical , Vascular Endothelial Growth Factor A/metabolism
6.
Gastroenterology ; 162(1): 179-192.e11, 2022 01.
Article in English | MEDLINE | ID: mdl-34425092

ABSTRACT

BACKGROUND AND AIMS: The enteric nervous system, which regulates many gastrointestinal functions, is derived from neural crest cells (NCCs). Defective NCC migration during embryonic development may lead to enteric neuropathies such as Hirschsprung's disease (hindgut aganglionosis). Sox10 is known to be essential for cell migration but downstream molecular events regulating early NCC migration have not been fully elucidated. This study aimed to determine how Sox10 regulates migration of sacral NCCs toward the hindgut using Dominant megacolon mice, an animal model of Hirschsprung's disease with a Sox10 mutation. METHODS: We used the following: time-lapse live cell imaging to determine the migration defects of mutant sacral NCCs; genome-wide microarrays, site-directed mutagenesis, and whole embryo culture to identify Sox10 targets; and liquid chromatography and tandem mass spectrometry to ascertain downstream effectors of Sox10. RESULTS: Sacral NCCs exhibited retarded migration to the distal hindgut in Sox10-null embryos with simultaneous down-regulated expression of cadherin-19 (Cdh19). Sox10 was found to bind directly to the Cdh19 promoter. Cdh19 knockdown resulted in retarded sacral NCC migration in vitro and ex vivo, whereas re-expression of Cdh19 partially rescued the retarded migration of mutant sacral NCCs in vitro. Cdh19 formed cadherin-catenin complexes, which then bound to filamentous actin of the cytoskeleton during cell migration. CONCLUSIONS: Cdh19 is a direct target of Sox10 during early sacral NCC migration toward the hindgut and forms cadherin-catenin complexes which interact with the cytoskeleton in migrating cells. Elucidation of this novel molecular pathway helps to provide insights into the pathogenesis of enteric nervous system developmental defects.


Subject(s)
Cadherins/metabolism , Cell Movement , Enteric Nervous System/metabolism , Hirschsprung Disease/metabolism , Neural Crest/metabolism , Neural Stem Cells/metabolism , Neurogenesis , SOXE Transcription Factors/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Cadherins/genetics , Cells, Cultured , Disease Models, Animal , Embryo Culture Techniques , Enteric Nervous System/abnormalities , Gene Expression Regulation, Developmental , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Neural Crest/abnormalities , Neural Stem Cells/pathology , Protein Binding , SOXE Transcription Factors/genetics , Signal Transduction , Time Factors
7.
Int J Mol Sci ; 22(20)2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34681906

ABSTRACT

The most common ventricular premature contractions (VPCs) originate from the right ventricular outflow tract (RVOT), but the molecular mechanisms of altered cytoskeletons of VPC-induced cardiomyopathy remain unexplored. We created a RVOT bigeminy VPC pig model (n = 6 in each group). Echocardiography was performed. The histopathological alternations in the LV myocardium were analyzed, and next generation sequencing (NGS) and functional enrichment analyses were employed to identify the differentially expressed genes (DEGs) responsible for the histopathological alternations. Finally, a cell silencing model was used to confirm the key regulatory gene and pathway. VPC pigs had increased LV diameters in the 6-month follow-up period. A histological study showed more actin cytoskeleton disorganization and actin accumulation over intercalated disc, Z-line arrangement disarray, increased ß-catenin expression, and cardiomyocyte enlargement in the LV myocardium of the VPC pigs compared to the control pigs. The NGS study showed actin cytoskeleton signaling, RhoGDI signaling, and signaling by Rho Family GTPases and ILK Signaling presented z-scores with same activation states. The expressions of Rac family small GTPase 2 (Rac2), the p-cofilin/cofilin ratio, and the F-actin/G-actin ratio were downregulated in the VPC group compared to the control group. Moreover, the intensity and number of actin filaments per cardiomyocyte were significantly decreased by Rac2 siRNA in the cell silencing model. Therefore, the Rac2/cofilin pathway was found to play a crucial role in the sarcomere morphology and Z-line arrangement disarray induced by RVOT bigeminy VPCs.


Subject(s)
Actin Cytoskeleton/pathology , Actin Depolymerizing Factors/metabolism , Arrhythmias, Cardiac/pathology , Heart Ventricles/pathology , Sarcomeres/pathology , rac GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/genetics , Animals , Arrhythmias, Cardiac/metabolism , Heart Ventricles/metabolism , Male , Sarcomeres/metabolism , Swine , Swine, Miniature , rac GTP-Binding Proteins/genetics , RAC2 GTP-Binding Protein
8.
PLoS One ; 16(5): e0248197, 2021.
Article in English | MEDLINE | ID: mdl-34038442

ABSTRACT

Canonical Wnt/ß-catenin (cWnt) signaling is a crucial regulator of development and Dishevelled (Dsh/Dvl) functions as an integral part of this pathway by linking Wnt binding to the Frizzled:LRP5/6 receptor complex with ß-catenin-stimulated gene expression. In many cell types Dsh has been localized to ill-defined cytoplasmic puncta, however in sea urchin eggs and embryos confocal fluorescence microscopy has shown that Dsh is localized to puncta present in a novel and development-essential vegetal cortex domain (VCD). In the present study, we used super-resolution light microscopy and platinum replica transmission electron microscopy (TEM) to provide the first views of the ultrastructural organization of Dsh within the sea urchin VCD. 3D structured illumination microscopy (SIM) imaging of isolated egg cortices demonstrated the graded distribution of Dsh in the VCD, whereas higher resolution stimulated emission depletion (STED) imaging revealed that some individual Dsh puncta consisted of more than one fluorescent source. Platinum replica immuno-TEM localization showed that Dsh puncta on the cytoplasmic face of the plasma membrane consisted of aggregates of pedestal-like structures each individually labeled with the C-terminus specific Dsh antibody. These aggregates were resistant to detergent extraction and treatment with drugs that disrupt actin filaments or inhibit myosin II contraction, and coexisted with the first cleavage actomyosin contractile ring. These results confirm and extend previous studies and reveal, for the first time in any cell type, the nanoscale organization of plasma membrane tethered Dsh. Our current working hypothesis is that these Dsh pedestals represent a prepositioned scaffold organization that is important for the localized activation of the cWnt pathway at the sea urchin vegetal pole. These observations in sea urchins may also be relevant to the submembranous Dsh puncta present in other eggs and embryos.


Subject(s)
Dishevelled Proteins/metabolism , Sea Urchins/metabolism , Wnt Signaling Pathway/physiology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Body Patterning , Dishevelled Proteins/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Microscopy, Electron, Transmission/methods , Myosin Type II/metabolism , Ovum/metabolism , Ovum/ultrastructure , Sea Urchins/growth & development
9.
Int J Mol Sci ; 22(4)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673054

ABSTRACT

There is ample evidence that, instead of a binary switch, epithelial-mesenchymal transition (EMT) in cancer results in a flexible array of phenotypes, each one uniquely suited to a stage in the invasion-metastasis cascade. The phenotypic plasticity of epithelium-derived cancer cells gives them an edge in surviving and thriving in alien environments. This review describes in detail the actin cytoskeleton and E-cadherin-based adherens junction rearrangements that cancer cells need to implement in order to achieve the advantageous epithelial/mesenchymal phenotype and plasticity of migratory phenotypes that can arise from partial EMT.


Subject(s)
Actin Cytoskeleton/metabolism , Adherens Junctions/metabolism , Epithelial-Mesenchymal Transition , Neoplasms/metabolism , Actin Cytoskeleton/pathology , Adherens Junctions/pathology , Humans , Neoplasms/pathology
10.
Toxins (Basel) ; 13(3)2021 02 28.
Article in English | MEDLINE | ID: mdl-33670922

ABSTRACT

The fungal metabolite sporidesmin is responsible for severe necrotizing inflammation of biliary tract and liver of livestock grazing on pasture containing spores of Pithomyces chartarum that synthesizes the toxin. The toxin is secreted into bile causing the erosion of the biliary epithelium accompanied by inflammation and damage to surrounding tissues. Toxicity has been suggested to be due to cycles of reduction and oxidation of sporidesmin leading to oxidative damage from the formation of reactive oxygen species. The current work is the first test of the oxidative stress hypothesis using cultured cells. Oxidative stress could not be detected in HepG2 cells incubated with sporidesmin using a dichlorodihydrofluorescein diacetate assay or by use of two-dimensional electrophoresis to search for oxidized peroxiredoxins. There was also no evidence for necrosis or apoptosis, although there was a loss of cell adhesion that was accompanied by the disruption of intracellular actin microfilaments that have known roles in cell adhesion. The results are consistent with a model in which altered contact between cells in situ leads to altered permeability and subsequent inflammation and necrosis, potentially from the leakage of toxic bile into surrounding tissues. There is now a need for the further characterization of the damage processes in vivo, including the investigation of altered permeability and mechanisms of cell death in the biliary tract and other affected organs.


Subject(s)
Actin Cytoskeleton/drug effects , Cell Adhesion/drug effects , Hepatocytes/drug effects , Sporidesmins/toxicity , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Apoptosis/drug effects , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Necrosis , Oxidative Stress/drug effects
11.
Arch Virol ; 166(5): 1371-1383, 2021 May.
Article in English | MEDLINE | ID: mdl-33715038

ABSTRACT

Herpesviruses are capable of infecting not only neurons, where they establish latent infection, but also astrocytes. Since astrocytes are important for the functioning of the central nervous system (CNS), their infection may lead to serious neurological disorders. Thus, in the present study we investigated the ability of human herpesvirus type 2 (HHV-2) to infect primary murine astrocytes in vitro and the effect of infection on their mitochondrial network and actin cytoskeleton. In immunofluorescence assays, antibodies against HHV-2 antigens and glial fibrillary acidic protein (GFAP) were used to confirm that the infected cells are indeed astrocytes. Real-time PCR analysis showed a high level of HHV-2 replication in astrocytes, particularly at 168 h postinfection, confirming that a productive infection had occurred. Analysis of mitochondrial morphology showed that, starting from the first stage of infection, HHV-2 caused fragmentation of the mitochondrial network and formation of punctate and tubular structures that colocalized with virus particles. Furthermore, during the late stages of infection, the infection affected the actin cytoskeleton and induced formation of actin-based cellular projections, which were probably associated with enhanced intracellular spread of the virus. These results suggest that the observed changes in the mitochondrial network and actin cytoskeleton in productively infected astrocytes are required for effective replication and viral spread in a primary culture of astrocytes. Moreover, we speculate that, in response to injury such as HHV-2 infection, murine astrocytes cultured in vitro undergo transformation, defined in vivo as reactive astrocytosis.


Subject(s)
Actin Cytoskeleton/pathology , Astrocytes/virology , Herpesvirus 2, Human/physiology , Mitochondria/pathology , Actin Cytoskeleton/metabolism , Animals , Astrocytes/pathology , Cells, Cultured , Gliosis , Kinetics , Mice , Mitochondria/metabolism , Virion/metabolism , Virus Replication
12.
Sci Rep ; 11(1): 3096, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542384

ABSTRACT

In recent years, there has been increasing interest in investigating the mechanical properties of individual cells to delineate disease mechanisms. Reorganization of cytoskeleton facilitates the colonization of metastatic breast cancer at bone marrow space, leading to bone metastasis. Here, we report evaluation of mechanical properties of two breast cancer cells with different metastatic ability at the site of bone metastases, using quasi-static and dynamic nanoindentation methods. Our results showed that the significant reduction in elastic modulus along with increased liquid-like behavior of bone metastasized MCF-7 cells was induced by depolymerization and reorganization of F-actin to the adherens junctions, whereas bone metastasized MDA-MB-231 cells showed insignificant changes in elastic modulus and F-actin reorganization over time, compared to their respective as-received counterparts. Taken together, our data demonstrate evolution of breast cancer cell mechanics at bone metastases.


Subject(s)
Actins/metabolism , Bone Neoplasms/pathology , Breast Neoplasms/pathology , Elastic Modulus/physiology , Actin Cytoskeleton/pathology , Actins/chemistry , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/secondary , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Breast Neoplasms/diagnostic imaging , Cytoskeleton/chemistry , Cytoskeleton/pathology , Female , Humans , MCF-7 Cells
13.
Eur J Pharmacol ; 895: 173866, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33454376

ABSTRACT

Metastatic breast cancer is a significant contributor to mortality among women, but its complex regulation represents a barrier to precision targeting. In the present study, a graphene-based nanocomposite which probes and selectively inhibits cancer cell motility is described. By controllable coupling of prenylated chalcone xanthohumol, an efficient inhibitor of mitochondrial electron transport chain complex I, with PEGylated graphene oxide nanosheet, a PEG-GO@XN nanocomposite with good stability and biocompatibility is synthesized. PEG-GO@XN is capable of inhibiting mitochondrial oxidative phosphorylation selectively in MDA-MB-231 and MDA-MB-436 metastatic breast cancer cells. PEG-GO@XN reduces the production of ATP, impairs the formation of F-actin cytoskeleton in the lamellipodia, and blocks the migration and invasion of breast cancer cells in vitro, without interfering the proliferation and metabolism of non-cancerous cells. More importantly, PEG-GO@XN suppresses the metastasis of MDA-MB-231 cells to lung in nude mice. PEG-GO@XN abolishes the TGF-ß1-induced down-regulation of E-cadherin and up-regulation of N-cadherin, vimentin, Snail and Twist, thus causes the maintenance of "epithelial-like" rather than the "mesenchymal-like" features, and decreases the motility potential of breast cancer cells. Taken together, this research unveils the enormous potential of PEG-GO@XN to suppress metastatic breast cancer by selective targeting oxidative phosphorylation and epithelial-mesenchymal transition of cancer cells and thereby providing insights on metastatic cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/prevention & control , Mitochondria/drug effects , Nanocomposites , Oxidative Phosphorylation/drug effects , Polyethylene Glycols/pharmacology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Adenosine Triphosphate/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Drug Compounding , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Mitochondria/pathology , Neoplasm Invasiveness , Pseudopodia/drug effects , Pseudopodia/metabolism , Pseudopodia/pathology , Signal Transduction , Xenograft Model Antitumor Assays
14.
PLoS One ; 16(1): e0246138, 2021.
Article in English | MEDLINE | ID: mdl-33508018

ABSTRACT

Visualizing actin filaments in fixed cells is of great interest for a variety of topics in cell biology such as cell division, cell movement, and cell signaling. We investigated the possibility of replacing phalloidin, the standard reagent for super-resolution imaging of F-actin in fixed cells, with the actin binding peptide 'lifeact'. We compared the labels for use in single molecule based super-resolution microscopy, where AlexaFluor 647 labeled phalloidin was used in a dSTORM modality and Atto 655 labeled lifeact was used in a single molecule imaging, reversible binding modality. We found that imaging with lifeact had a comparable resolution in reconstructed images and provided several advantages over phalloidin including lower costs, the ability to image multiple regions of interest on a coverslip without degradation, simplified sequential super-resolution imaging, and more continuous labeling of thin filaments.


Subject(s)
Actin Cytoskeleton/pathology , Carbocyanines/chemistry , Phalloidine/chemistry , Actin Cytoskeleton/chemistry , HeLa Cells , Humans , Microscopy, Fluorescence
15.
Small GTPases ; 12(3): 202-208, 2021 05.
Article in English | MEDLINE | ID: mdl-31648598

ABSTRACT

Oncogenic protein kinase C epsilon (PKCε) promotes the formation of membrane ruffles and motility in non-small cell lung cancer (NSCLC) cells. We found that PKCε is down-regulated when NSCLC cells undergo epithelial-to-mesenchymal transition (EMT) in response to TGF-ß, thus becoming dispensable for migration and invasion in the mesenchymal state. PKCε silencing or inhibition leads to stress fibre formation, suggesting that this kinase negatively regulates RhoA activity. Ruffle formation induced by PKCε activation in the epithelial state is dependent on PI3K, but does not involve the PI3K-dependent Rac-GEFs Ect2, Trio, Vav2 or Tiam1, suggesting alternative Rac-GEFs as mediators of this response. In the proposed model, PKCε acts as a rheostat for Rho GTPases that differs in the epithelial and mesenchymal states.


Subject(s)
Actin Cytoskeleton/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Protein Kinase C-epsilon/metabolism , rho GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Kinase C-epsilon/genetics , Signal Transduction , Tumor Cells, Cultured , rho GTP-Binding Proteins/genetics
16.
FEBS J ; 288(10): 3120-3134, 2021 05.
Article in English | MEDLINE | ID: mdl-32885587

ABSTRACT

Serum response factor (SRF), a member of the Mcm1, Agamous, Deficiens, and SRF (MADS) box transcription factor, is widely expressed in all cell types and plays a crucial role in the physiological function and development of diseases. SRF regulates its downstream genes by binding to their CArG DNA box by interacting with various cofactors. However, the underlying mechanisms are not fully understood, therefore attracting increasing research attention due to the importance of this topic. This review's objective is to discuss the new progress in the studies of the molecular mechanisms involved in the activation of SRF and its impacts in physiological and pathological conditions. Notably, we summarized the recent studies on the interaction of SRF with its two main types of cofactors belonging to the myocardin families of transcription factors and the members of the ternary complex factors. The knowledge of these mechanisms will create new opportunities for understanding the dynamics of many traits and disease pathogenesis especially, cardiovascular diseases and cancer that could serve as targets for pharmacological control and treatment of these diseases.


Subject(s)
Cardiovascular Diseases/genetics , Neoplasms/genetics , Nuclear Proteins/genetics , Serum Response Factor/genetics , Ternary Complex Factors/genetics , Trans-Activators/genetics , Transcription, Genetic , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Apoptosis/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cell Proliferation , DNA/genetics , DNA/metabolism , Gene Expression Regulation , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Serum Response Factor/metabolism , Signal Transduction , Stress, Physiological , Ternary Complex Factors/metabolism , Trans-Activators/metabolism
17.
Am J Physiol Renal Physiol ; 320(1): F97-F113, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33308016

ABSTRACT

We recently reported that the enhanced susceptibility to chronic kidney disease (CKD) in the fawn-hooded hypertensive (FHH) rat is caused, at least in part, by a mutation in γ-adducin (ADD3) that attenuates renal vascular function. The present study explored whether Add3 contributes to the modulation of podocyte structure and function using FHH and FHH.Add3 transgenic rats. The expression of ADD3 on the membrane of primary podocytes isolated from FHH was reduced compared with FHH.Add3 transgenic rats. We found that F-actin nets, which are typically localized in the lamellipodia, replaced unbranched stress fibers in conditionally immortalized mouse podocytes transfected with Add3 Dicer-substrate short interfering RNA (DsiRNA) and primary podocytes isolated from FHH rats. There were increased F/G-actin ratios and expression of the Arp2/3 complexes throughout FHH podocytes in association with reduced synaptopodin and RhoA but enhanced Rac1 and CDC42 expression in the renal cortex, glomeruli, and podocytes of FHH rats. The expression of nephrin at the slit diaphragm and the levels of focal adhesion proteins integrin-α3 and integrin-ß1 were decreased in the glomeruli of FHH rats. Cell migration was enhanced and adhesion was reduced in podocytes of FHH rats as well as in immortalized mouse podocytes transfected with Add3 DsiRNA. Mean arterial pressures were similar in FHH and FHH.Add3 transgenic rats at 16 wk of age; however, FHH rats exhibited enhanced proteinuria associated with podocyte foot process effacement. These results demonstrate that reduced ADD3 function in FHH rats alters baseline podocyte pathophysiology by rearrangement of the actin cytoskeleton at the onset of proteinuria in young animals.


Subject(s)
Actin Cytoskeleton/metabolism , Calmodulin-Binding Proteins/metabolism , Hypertension/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Renal Insufficiency, Chronic/metabolism , Actin Cytoskeleton/pathology , Animals , Arterial Pressure , Calmodulin-Binding Proteins/genetics , Cell Adhesion , Cell Line , Cell Movement , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Disease Progression , Focal Adhesions/metabolism , Focal Adhesions/pathology , Hypertension/genetics , Hypertension/pathology , Hypertension/physiopathology , Integrins/metabolism , Male , Mice , Monomeric GTP-Binding Proteins/metabolism , Podocytes/pathology , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/physiopathology , Rats, Inbred Strains , Rats, Transgenic , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Signal Transduction
18.
Biomed Pharmacother ; 132: 110883, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33113417

ABSTRACT

Curcumin is a turmeric, antioxidative compound, well-known of its anti-cancer properties. Nowadays more and more effort is made in the field of enhancing the efficiency of the anticancer therapies. Combining the photoactive properties of curcumin with the superficial localization of melanoma and photodynamic therapy (PDT) seems to be a promising treatment method. The research focused on the evaluation of the curcumin effectiveness as an anticancer therapeutic agent in the in vitro treatment of melanotic (A375) and amelanotic (C32) melanoma cell lines. Keratinocytes (HaCat) and fibroblasts (HGF) were used to assess the impact of the therapy on the skin tissue. The aim of the study was to investigate the cell death after exposure to light irradiation after preincubation with curcumin. Additionaly the authors analized the interactions between curcumin and the actin cytoskeleton. The cytotoxic effect initiated by curcumin and increased by irradiation confirm the usefulness of the flavonoid in the PDT approach. Depending on curcumin concentration and incubation time, melanoma cells survival rate ranged from: 93.68 % (C32 cell line, 10 µM, 24 h) and 83.47 % (A375 cell line, 10 µM, 24 h) to 8.98 % (C32 cell line, 50 µM, 48 h) and 12.42 % (A375 cell line, 50 µM, 48 h). Moreover, photodynamic therapy with curcumin increased the number of apoptotic and necrotic cells in comparison to incubation with curcumin without irradiation. The study demonstrated that PDT induced caspase-3 overexpression and DNA cleavage in the studied cell lines. The cells revealed decreased proliferation after the therapy due to the actin cytoskeleton rearrangement. Although effective, the therapy remains not selective towards melanoma cells.


Subject(s)
Actin Cytoskeleton/drug effects , Curcumin/pharmacology , Melanins/metabolism , Melanocytes/drug effects , Melanoma/drug therapy , Photochemotherapy , Photosensitizing Agents/pharmacology , Skin Neoplasms/drug therapy , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Male , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Middle Aged , Necrosis , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
19.
Int Rev Cell Mol Biol ; 356: 131-196, 2020.
Article in English | MEDLINE | ID: mdl-33066873

ABSTRACT

The cytoskeleton is a central factor contributing to various hallmarks of cancer. In recent years, there has been increasing evidence demonstrating the involvement of actin regulatory proteins in malignancy, and their dysregulation was shown to predict poor clinical prognosis. Although enhanced cytoskeletal activity is often associated with cancer progression, the expression of several inducers of actin polymerization is remarkably reduced in certain malignancies, and it is not completely clear how these changes promote tumorigenesis and metastases. The complexities involved in cytoskeletal induction of cancer progression therefore pose considerable difficulties for therapeutic intervention; it is not always clear which cytoskeletal regulator should be targeted in order to impede cancer progression, and whether this targeting may inadvertently enhance alternative invasive pathways which can aggravate tumor growth. The entire constellation of cytoskeletal machineries in eukaryotic cells are numerous and complex; the system is comprised of and regulated by hundreds of proteins, which could not be covered in a single review. Therefore, we will focus here on the actin cytoskeleton, which encompasses the biological machinery behind most of the key cellular functions altered in cancer, with specific emphasis on actin nucleating factors and nucleation-promoting factors. Finally, we discuss current therapeutic strategies for cancer which aim to target the cytoskeleton.


Subject(s)
Actin Cytoskeleton/immunology , Actins/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Signal Transduction/immunology , Actin Cytoskeleton/pathology , Animals , Disease Progression , Humans , Neoplasm Metastasis , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...