Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 691
Filter
1.
Theranostics ; 12(2): 782-795, 2022.
Article in English | MEDLINE | ID: mdl-34976213

ABSTRACT

Rationale: Polycystic ovary syndrome (PCOS) is closely linked to follicular dysplasia and impaired bidirectional oocyte-granulosa cell (GC) communication. Given that PCOS is a heterogeneous, multifactorial endocrine disorder, it is important to clarify the pathophysiology of this ovarian disease and identify a specific treatment. Methods: We generated PCOS rat models based on neonatal tributyltin (TBT) exposure and studied the therapeutic effect and mechanism of resveratrol (RSV), a natural plant polyphenol. Transcriptome analysis was conducted to screen the significantly changed pathways, and a series of experiments, such as quantitative real-time polymerase chain reaction (PCR), Western blot and phalloidin staining, were performed in rat ovaries. We also observed similar changes in human PCOS samples using Gene Expression Omnibus (GEO) database analysis and quantitative real-time PCR. Results: We first found that injury to transzonal projections (TZPs), which are specialized filopodia that mediate oocyte-GC communication in follicles, may play an important role in the etiology of PCOS. We successfully established PCOS rat models using TBT and found that overexpressed calcium-/calmodulin-dependent protein kinase II beta (CaMKIIß) inhibited TZP assembly. In addition, TZP disruption and CAMK2B upregulation were also observed in samples from PCOS patients. Moreover, we demonstrated that RSV potently ameliorated ovarian failure and estrus cycle disorder through TZP recovery via increased cytoplasmic calcium levels and excessive phosphorylation of CaMKIIß. Conclusions: Our data indicated that upregulation of CaMKIIß may play a critical role in regulating TZP assembly and may be involved in the pathogenesis of PCOS associated with ovarian dysfunction. Investigation of TZPs and RSV as potent CaMKIIß activators provides new insight and a therapeutic target for PCOS, which is helpful for improving female reproduction.


Subject(s)
Cell Communication/drug effects , Granulosa Cells/drug effects , Oocytes/drug effects , Polycystic Ovary Syndrome/drug therapy , Pseudopodia/drug effects , Resveratrol/therapeutic use , Adult , Animals , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Disease Models, Animal , Female , Granulosa Cells/metabolism , Humans , Oocytes/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Pseudopodia/metabolism , Rats , Rats, Sprague-Dawley , Trialkyltin Compounds
2.
Mol Biol Cell ; 33(1): ar9, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34788129

ABSTRACT

The ability of cells to sense chemical gradients is essential during development, morphogenesis, and immune responses. Although much is known about chemoattraction, chemorepulsion remains poorly understood. Proliferating Dictyostelium cells secrete a chemorepellent protein called AprA. AprA prevents pseudopod formation at the region of the cell closest to the source of AprA, causing the random movement of cells to be biased away from the AprA. Activation of Ras proteins in a localized sector of a cell cortex helps to induce pseudopod formation, and Ras proteins are needed for AprA chemorepulsion. Here we show that AprA locally inhibits Ras cortical activation through the G protein-coupled receptor GrlH, the G protein subunits Gß and Gα8, Ras protein RasG, protein kinase B, the p21-activated kinase PakD, and the extracellular signal-regulated kinase Erk1. Diffusion calculations and experiments indicate that in a colony of cells, high extracellular concentrations of AprA in the center can globally inhibit Ras activation, while a gradient of AprA that naturally forms at the edge of the colony allows cells to activate Ras at sectors of the cell other than the sector of the cell closest to the center of the colony, effectively inducing both repulsion from the colony and cell differentiation. Together, these results suggest that a pathway that inhibits local Ras activation can mediate chemorepulsion.


Subject(s)
Cell Migration Inhibition/physiology , Dictyostelium/drug effects , Dictyostelium/metabolism , Cell Migration Inhibition/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemotaxis/drug effects , Chemotaxis/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Protozoan Proteins/metabolism , Pseudopodia/drug effects , Pseudopodia/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , p21-Activated Kinases/metabolism , ras Proteins/metabolism
3.
Cells ; 10(12)2021 11 26.
Article in English | MEDLINE | ID: mdl-34943828

ABSTRACT

Drosophila suzukii is a neobiotic invasive pest that causes extensive damage to fruit crops worldwide. The biological control of this species has been unsuccessful thus far, in part because of its robust cellular innate immune system, including the activity of professional phagocytes known as hemocytes and plasmatocytes. The in vitro cultivation of primary hemocytes isolated from D. suzukii third-instar larvae is a valuable tool for the investigation of hemocyte-derived effector mechanisms against pathogens such as wasp parasitoid larvae, bacteria, fungi and viruses. Here, we describe the morphological characteristics of D. suzukii hemocytes and evaluate early innate immune responses, including extracellular traps released against the entomopathogen Pseudomonas entomophila and lipopolysaccharides. We show for the first time that D. suzukii plasmatocytes cast extracellular traps to combat P. entomophila, along with other cell-mediated reactions, such as phagocytosis and the formation of filopodia.


Subject(s)
Drosophila/immunology , Drosophila/microbiology , Extracellular Traps/metabolism , Immunity, Innate , Introduced Species , Pseudomonas/physiology , Animals , Cell Survival/drug effects , Drosophila/ultrastructure , Extracellular Traps/drug effects , Hemocytes/drug effects , Hemocytes/ultrastructure , Immunity, Innate/drug effects , Larva/cytology , Lipopolysaccharides/pharmacology , Phagocytes/drug effects , Phagocytes/microbiology , Pseudomonas/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism
4.
Cells ; 10(12)2021 12 10.
Article in English | MEDLINE | ID: mdl-34943993

ABSTRACT

The lamellipodia and pseudopodia of migrating cells are produced and maintained by the Scar/WAVE complex. Thus, actin-based cell migration is largely controlled through regulation of Scar/WAVE. Here, we report that the Abi subunit-but not Scar-is phosphorylated in response to extracellular signalling in Dictyostelium cells. Like Scar, Abi is phosphorylated after the complex has been activated, implying that Abi phosphorylation modulates pseudopodia, rather than causing new ones to be made. Consistent with this, Scar complex mutants that cannot bind Rac are also not phosphorylated. Several environmental cues also affect Abi phosphorylation-cell-substrate adhesion promotes it and increased extracellular osmolarity diminishes it. Both unphosphorylatable and phosphomimetic Abi efficiently rescue the chemotaxis of Abi KO cells and pseudopodia formation, confirming that Abi phosphorylation is not required for activation or inactivation of the Scar/WAVE complex. However, pseudopodia and Scar patches in the cells with unphosphorylatable Abi protrude for longer, altering pseudopod dynamics and cell speed. Dictyostelium, in which Scar and Abi are both unphosphorylatable, can still form pseudopods, but migrate substantially faster. We conclude that extracellular signals and environmental responses modulate cell migration by tuning the behaviour of the Scar/WAVE complex after it has been activated.


Subject(s)
Dictyostelium/metabolism , Extracellular Space/metabolism , Protozoan Proteins/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Chemotactic Factors/pharmacology , Dictyostelium/drug effects , Mutation/genetics , Osmotic Pressure/drug effects , Phosphorylation/drug effects , Protozoan Proteins/genetics , Pseudopodia/drug effects , Pseudopodia/metabolism , Signal Transduction/drug effects
5.
Cells ; 10(11)2021 10 28.
Article in English | MEDLINE | ID: mdl-34831155

ABSTRACT

Platelets are functionally versatile blood cells involved in thrombosis, hemostasis, atherosclerosis, and immune response. Platelet interaction with the immediate microenvironment in blood, vasculature, and tissues alters platelet morphology. The quantification of platelet morphodynamics by geometrical parameters (morphometry) can provide important insights into how platelets sense and respond to stimulatory cues in their vicinity. However, the extraction of platelet shapes from phase contrast microscopy images by conventional image processing is difficult. Here, we used a convolutional neural network (CNN) to develop a deep-learning-based approach for the unbiased extraction of information on platelet morphodynamics by phase contrast microscopy. We then investigated the effect of normal and oxidized low-density lipoproteins (LDL, oxLDL) on platelet morphodynamics, spreading, and haptotactic migration. Exposure of platelets to oxLDL led to a decreased spreading area and rate on fibrinogen, accompanied by increased formation of filopodia and impaired formation of lamellipodia. Haptotactic platelet migration was affected by both LDL and oxLDL in terms of decreased migration velocity and reduced directional persistence. Our results demonstrate the use of deep learning in investigating platelet morphodynamics and reveal differential effects of LDL and oxLDL on platelet morphology and platelet-matrix interaction.


Subject(s)
Blood Platelets/cytology , Cell Movement , Cell Shape , Deep Learning , Lipoproteins, LDL/pharmacology , Cell Movement/drug effects , Cell Shape/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Pseudopodia/drug effects , Pseudopodia/metabolism , Touch
6.
ACS Appl Mater Interfaces ; 13(41): 49542-49555, 2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34610736

ABSTRACT

To solve the poor sustainability of electroactive stimulation in clinical therapy, a strategy of combining a piezoelectric BaTiO3-coated Ti6Al4V scaffold and low-intensity pulsed ultrasound (LIPUS) was unveiled and named here as piezodynamic therapy. Thus, cell behavior could be regulated phenomenally by force and electricity simultaneously. First, BaTiO3 was deposited uniformly on the surface of the three-dimensional (3D) printed porous Ti6Al4V scaffold, which endowed the scaffold with excellent force-electricity responsiveness under pulsed ultrasound exposure. The results of live/dead staining, cell scanning electron microscopy, and F-actin staining showed that cells had better viability, better pseudo-foot adhesion, and more muscular actin bundles when they underwent the piezodynamic effect of ultrasound and piezoelectric coating. This piezodynamic therapy activated more mitochondria at the initial stage that intervened in the cell cycle by promoting cells' proliferation and weakened the apoptotic damage. The quantitative real-time polymerase chain reaction data further confirmed that the costimulation of the ultrasound and the piezoelectric scaffolds could trigger adequate current to upregulated the expression of osteogenic-related genes. The continuous electric cues could be generated by the BaTiO3-coated scaffold and intermittent LIPUS stimulation; thereon, more efficient bone healing would be promoted by piezodynamic therapy in future treatment.


Subject(s)
Alloys/chemistry , Barium Compounds/chemistry , Tissue Scaffolds/chemistry , Titanium/chemistry , Ultrasonic Waves , Alloys/radiation effects , Animals , Apoptosis/drug effects , Barium Compounds/radiation effects , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Membrane Potential, Mitochondrial/drug effects , Mesenchymal Stem Cells/drug effects , Mitochondria/drug effects , Osteogenesis/drug effects , Porosity , Pseudopodia/drug effects , Rats, Sprague-Dawley , Titanium/radiation effects , Wettability
7.
Exp Cell Res ; 407(2): 112753, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34499887

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) activation has been reported to exert protective effects on podocytes, whereas angiopoietin-like 3 (ANGPTL3) has been shown to exert significant pathogenic effects on these cells. This study aimed to investigate the link between the protective effects of PPARα activation and the pathogenic effects of ANGPTL3 in podocytes. Both PPARα and ANGPTL3 were expressed in cultured podocytes. PPARα mRNA and protein levels decreased whereas ANGPTL3 mRNA and protein levels increased in a time-dependent manner in podocytes treated with puromycin aminonucleoside (PAN). Gemfibrozil, a pharmacological agonist of PPARα, increased PPARα levels and activity in podocytes. The drug also decreased ANGPTL3 levels by potentially weakening ANGPTL3 promoter activity in both normal and PAN-treated podocytes. Furthermore, gemfibrozil significantly decreased PAN-induced apoptosis and F-actin rearrangement. Primary podocytes from Angptl3-knockout mice were cultured. There was no significant difference between Angptl3-/- podocytes treated with or without gemfibrozil in the lamellipodia numbers after PAN treatment. The results suggested that the protective effects of gemfibrozil on podocytes were not exerted following knockout of the Angptl3 gene. This study identified a novel mechanism of the PPARα agonist gemfibrozil that exerts its protective effects by inhibiting PAN-induced apoptosis and cytoskeleton rearrangements through inhibition of ANGPTL3 expression.


Subject(s)
Actin Cytoskeleton/drug effects , Angiopoietin-like Proteins/physiology , Gemfibrozil/pharmacology , PPAR alpha/agonists , Podocytes/drug effects , Pseudopodia/drug effects , Puromycin Aminonucleoside/pharmacology , Angiopoietin-Like Protein 3 , Animals , Apoptosis , Hypolipidemic Agents/pharmacology , Mice , Mice, Knockout , Podocytes/metabolism , Podocytes/pathology , Protective Factors , Pseudopodia/metabolism
8.
PLoS One ; 16(8): e0254929, 2021.
Article in English | MEDLINE | ID: mdl-34383763

ABSTRACT

In searching for novel targeted therapeutic agents for lung cancer treatment, norcycloartocarpin from Artocarpus gomezianus was reported in this study to promisingly interacted with Akt and exerted the apoptosis induction and epithelial-to-mesenchymal transition suppression. Selective cytotoxic profile of norcycloartocarpin was evidenced with approximately 2-fold higher IC50 in normal dermal papilla cells (DPCs) compared with human lung cancer A549, H460, H23, and H292 cells. We found that norcycloartocarpin suppressed anchorage-independent growth, cell migration, invasion, filopodia formation, and decreased EMT in a dose-dependent manner at 24 h, which were correlated with reduced protein levels of N-cadherin, Vimentin, Slug, p-FAK, p-Akt, as well as Cdc42. In addition, norcycloartocarpin activated apoptosis caspase cascade associating with restoration of p53, down-regulated Bcl-2 and augmented Bax in A549 and H460 cells. Interestingly, norcycloartocarpin showed potential inhibitory role on protein kinase B (Akt) the up-stream dominant molecule controlling EMT and apoptosis. Computational molecular docking analysis further confirmed that norcycloartocarpin has the best binding affinity of -12.52 kcal/mol with Akt protein at its critical active site. As Akt has recently recognized as an attractive molecular target for therapeutic approaches, these findings support its use as a plant-derived anticancer agent in cancer therapy.


Subject(s)
Epithelial-Mesenchymal Transition , Flavonoids/pharmacology , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Flavonoids/chemistry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Lung Neoplasms/drug therapy , Molecular Docking Simulation , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/chemistry , Pseudopodia/drug effects , Pseudopodia/metabolism , Signal Transduction/drug effects , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/metabolism
9.
PLoS Comput Biol ; 17(7): e1008803, 2021 07.
Article in English | MEDLINE | ID: mdl-34260581

ABSTRACT

During the last decade, a consensus has emerged that the stochastic triggering of an excitable system drives pseudopod formation and subsequent migration of amoeboid cells. The presence of chemoattractant stimuli alters the threshold for triggering this activity and can bias the direction of migration. Though noise plays an important role in these behaviors, mathematical models have typically ignored its origin and merely introduced it as an external signal into a series of reaction-diffusion equations. Here we consider a more realistic description based on a reaction-diffusion master equation formalism to implement these networks. In this scheme, noise arises naturally from a stochastic description of the various reaction and diffusion terms. Working on a three-dimensional geometry in which separate compartments are divided into a tetrahedral mesh, we implement a modular description of the system, consisting of G-protein coupled receptor signaling (GPCR), a local excitation-global inhibition mechanism (LEGI), and signal transduction excitable network (STEN). Our models implement detailed biochemical descriptions whenever this information is available, such as in the GPCR and G-protein interactions. In contrast, where the biochemical entities are less certain, such as the LEGI mechanism, we consider various possible schemes and highlight the differences between them. Our simulations show that even when the LEGI mechanism displays perfect adaptation in terms of the mean level of proteins, the variance shows a dose-dependence. This differs between the various models considered, suggesting a possible means for determining experimentally among the various potential networks. Overall, our simulations recreate temporal and spatial patterns observed experimentally in both wild-type and perturbed cells, providing further evidence for the excitable system paradigm. Moreover, because of the overall importance and ubiquity of the modules we consider, including GPCR signaling and adaptation, our results will be of interest beyond the field of directed migration.


Subject(s)
Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Computer Simulation , Models, Biological , Computational Biology , Diffusion , Pseudopodia/drug effects , Stochastic Processes
10.
Anticancer Res ; 41(8): 3843-3849, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34281844

ABSTRACT

BACKGROUND/AIM: Integrin-targeting compounds have shown clinically significant benefits in many patients. Here, we examined the activity of millettocalyxin B, extracted from the stem bark of Millettia erythrocalyx, in lung cancer cells. MATERIALS AND METHODS: The viability of human lung cancer cells was investigated by the 3-(4,5-dimethylthiazol-2-yl)-2,5diphenyl tetrazoliumbromide (MTT) assay. Migration and invasion assays were performed. Phalloidin-rhodamine staining was used to determine the formation of filopodia. Western blot analysis and immunofluorescence staining were used to identify the signaling proteins involved in migration regulation. RESULTS: Non-toxic concentrations (0-25 µM) of millettocalyxin B reduced migration and invasion of lung cancer A549 cells. Filopodia were significantly reduced in millettocalyxin B-treated cells. The migration regulatory proteins including integrin α5, active FAK, active Akt, and Cdc42 were significantly decreased in Millettocalyxin B-treated cells. CONCLUSION: Our findings revealed a novel anti-migration and anti-invasion effects and the underlying mechanism of millettocalyxin B, which may be exploited for cancer treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavonoids/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , A549 Cells , Cell Movement/drug effects , Cell Movement/physiology , Focal Adhesion Kinase 1/metabolism , Humans , Integrin alpha5/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pseudopodia/drug effects , cdc42 GTP-Binding Protein/metabolism
11.
Angew Chem Int Ed Engl ; 60(40): 21905-21910, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34322970

ABSTRACT

In solid tumors, tumor invasion and metastasis account for 90 % of cancer-related deaths. Cell migration is steered by the lamellipodia formed at the leading edge. These lamellipodia can drive the cell body forward by its mechanical deformation regulated by cofilin. Inhibiting cofilin activity can cause significant defects in directional lamellipodia formation and the locomotory capacity of cell invasion, thus contributing to antimetastatic treatment. Herein, a near infrared light (NIR)-controlled nanoscale proton supplier was designed with upconversion nanoparticles (UCNPs) as a core coated in MIL-88B for interior photoacids loading; this photoacids loading can boost H+ transients in cells, which converts the cofilin to an inactive form. Strikingly, inactive cofilin loses the ability to mediate lamellipodia deformation for cell migration. Additionally, the iron, which serves as a catalyticaly active center in MIL-88B, initiates an enhanced Fenton reaction due to the increased H+ in the tumor, ultimately achieving intensive chemodynamic therapy (CDT). This work provides new insight into H+ transients in cells, which not only regulates cofilin protonation for antimetastatic treatment but also improves chemodynamic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Metal-Organic Frameworks/pharmacology , Nanoparticles/chemistry , Photochemotherapy , Pseudopodia/drug effects , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Infrared Rays , Metal-Organic Frameworks/chemistry , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Particle Size , Surface Properties
12.
Aging (Albany NY) ; 13(13): 17901-17913, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34170850

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is characterized by a high rate of metastasis. It has been found that tumor cells can bypass apoptosis which leads to an uncontrolled proliferation, but chloroquine (CQ) can have an effect on the tumors by inducing apoptosis. We aimed to explore the effects and the hypothetical mechanism of CQ effects on OS. METHODS: We first estimated the CQ effects on proliferation, apoptosis, migration, invasion, and lamellipodia formation of OS cells. Mice bearing xenograft model were used to test the anti-tumor growth and lung metastasis effects of CQ in OS. Western blot and immunohistochemistry were used to explore the mechanism of CQ effects and the association between p-STAT3 expression and lung metastasis of OS patients. RESULTS: CQ induces the apoptosis and suppressed the viability, proliferation, migration, invasion, and lamellipodia formation of OS cells in vitro. In vivo experiments demonstrated that CQ inhibited tumor growth and lung metastasis. CQ induced apoptosis was dependent on the lysosomal inhibition and inhibition of protein turnover. The lung metastasis was associated with the p-STAT3 expression in OS patients. CONCLUSION: CQ inhibited progression of OS cells in vitro, and suppressed tumor growth and lung metastasis in vivo. p-STAT3 can be a predictive biomarker for lung metastasis in osteosarcoma patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Proliferation/drug effects , Chloroquine/pharmacology , Neoplasm Invasiveness/pathology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , STAT3 Transcription Factor/metabolism , Adult , Animals , Bone Neoplasms/metabolism , Female , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/prevention & control , Osteosarcoma/metabolism , Phosphorylation , Pseudopodia/drug effects , Xenograft Model Antitumor Assays , Young Adult
13.
Cell Adh Migr ; 15(1): 166-179, 2021 12.
Article in English | MEDLINE | ID: mdl-34152257

ABSTRACT

Microtubules (MTs) control cell shape and intracellular cargo transport. The role of MT turnover in the migration of slow-moving cells through endothelial barriers remains unclear. To irreversibly interfere with MT disassembly, we have used the MT-stabilizing agent zampanolide (ZMP) in Β16F10 melanoma as amodel of slow-moving cells. ZMP-treated B16 cells failed to follow chemotactic gradients across rigid confinements and could not generate stable sub-endothelial pseudopodia under endothelial monolayers. In vivo, ZMP-treated Β16 cells failed to extravasate though lung capillaries. In contrast to melanoma cells, the chemotaxis and transendothelial migration of ZMP-treated Tcells were largely conserved. This is afirst demonstration that MT disassembly is akey checkpoint in the directional migration of cancer cells but not of lymphocytes.


Subject(s)
Chemotaxis/drug effects , Macrolides/pharmacology , Microtubules/metabolism , Transendothelial and Transepithelial Migration/drug effects , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Male , Melanoma/pathology , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Pseudopodia/drug effects , T-Lymphocytes/drug effects
14.
J Biol Chem ; 297(1): 100819, 2021 07.
Article in English | MEDLINE | ID: mdl-34029590

ABSTRACT

Collagen-derived hydroxyproline (Hyp)-containing peptides have a variety of biological effects on cells. These bioactive collagen peptides are locally generated by the degradation of endogenous collagen in response to injury. However, no comprehensive study has yet explored the functional links between Hyp-containing peptides and cellular behavior. Here, we show that the dipeptide prolyl-4-hydroxyproline (Pro-Hyp) exhibits pronounced effects on mouse tendon cells. Pro-Hyp promotes differentiation/maturation of tendon cells with modulation of lineage-specific factors and induces significant chemotactic activity in vitro. In addition, Pro-Hyp has profound effects on cell proliferation, with significantly upregulated extracellular signal-regulated kinase phosphorylation and extracellular matrix production and increased type I collagen network organization. Using proteomics, we have predicted molecular transport, cellular assembly and organization, and cellular movement as potential linked-network pathways that could be altered in response to Pro-Hyp. Mechanistically, cells treated with Pro-Hyp demonstrate increased directional persistence and significantly increased directed motility and migration velocity. They are accompanied by elongated lamellipodial protrusions with increased levels of active ß1-integrin-containing focal contacts, as well as reorganization of thicker peripheral F-actin fibrils. Pro-Hyp-mediated chemotactic activity is significantly reduced (p < 0.001) in cells treated with the mitogen-activated protein kinase kinase 1/2 inhibitor PD98059 or the α5ß1-integrin antagonist ATN-161. Furthermore, ATN-161 significantly inhibits uptake of Pro-Hyp into adult tenocytes. Thus, our findings document the molecular basis of the functional benefits of the Pro-Hyp dipeptide in cellular behavior. These dynamic properties of collagen-derived Pro-Hyp dipeptide could lead the way to its application in translational medicine.


Subject(s)
Cell Movement/drug effects , Dipeptides/pharmacology , Homeostasis/drug effects , Integrin beta1/metabolism , Pseudopodia/metabolism , Tendons/cytology , Aging , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Pseudopodia/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Tenocytes/cytology , Tenocytes/drug effects , Up-Regulation/drug effects
15.
Cell Rep ; 35(2): 108971, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852848

ABSTRACT

In schizophrenia (SCZ), neurons in the brain tend to undergo gross morphological changes, but the related molecular mechanism remains largely elusive. Using Kif3b+/- mice as a model with SCZ-like behaviors, we found that a high-betaine diet can significantly alleviate schizophrenic traits related to neuronal morphogenesis and behaviors. According to a deficiency in the transport of collapsin response mediator protein 2 (CRMP2) by the KIF3 motor, we identified a significant reduction in lamellipodial dynamics in developing Kif3b+/- neurons as a cause of neurite hyperbranching. Betaine administration significantly decreases CRMP2 carbonylation, which enhances the F-actin bundling needed for proper lamellipodial dynamics and microtubule exclusion and may thus functionally compensate for KIF3 deficiency. Because the KIF3 expression levels tend to be downregulated in the human prefrontal cortex of the postmortem brains of SCZ patients, this mechanism may partly participate in human SCZ pathogenesis, which we hypothesize could be alleviated by betaine administration.


Subject(s)
Betaine/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Kinesins/genetics , Nerve Tissue Proteins/genetics , Neurons/drug effects , Prefrontal Cortex/drug effects , Pseudopodia/drug effects , Schizophrenia/diet therapy , Actins/genetics , Actins/metabolism , Animals , Behavior, Animal/drug effects , Biological Transport , Diet/methods , Disease Models, Animal , Gene Expression Regulation, Developmental , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Kinesins/deficiency , Male , Mice , Mice, Knockout , Microtubules/drug effects , Microtubules/metabolism , Microtubules/ultrastructure , Nerve Tissue Proteins/deficiency , Neurons/metabolism , Neurons/ultrastructure , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Protein Binding , Protein Carbonylation , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology
16.
Acta Pharmacol Sin ; 42(12): 2033-2045, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33664417

ABSTRACT

Caffeine induces multiple vascular effects. In this study we investigated the angiogenic effect of physiological concentrations of caffeine with focus on endothelial cell behaviors (migration and proliferation) during angiogenesis and its mitochondrial and bioenergetic mechanisms. We showed that caffeine (10-50 µM) significantly enhanced angiogenesis in vitro, evidenced by concentration-dependent increases in tube formation, and migration of human umbilical vein endothelial cells (HUVECs) without affecting cell proliferation. Caffeine (50 µM) enhanced endothelial migration via activation of cAMP/PKA/AMPK signaling pathway, which was mimicked by cAMP analog 8-Br-cAMP, and blocked by PKA inhibitor H89, adenylate cyclase inhibitor SQ22536 or AMPK inhibitor compound C. Furthermore, caffeine (50 µM) induced significant mitochondrial shortening through the increased phosphorylation of mitochondrial fission protein dynamin-related protein 1 (Drp1) in HUVECs, which increased its activity to regulate mitochondrial fission. Pharmacological blockade of Drp1 by Mdivi-1 (10 µM) or disturbance of mitochondrial fission by Drp1 silencing markedly suppressed caffeine-induced lamellipodia formation and endothelial cell migration. Moreover, we showed that caffeine-induced mitochondrial fission led to accumulation of more mitochondria in lamellipodia regions and augmentation of mitochondrial energetics, both of which were necessary for cell migration. In a mouse model of hindlimb ischemia, administration of caffeine (0.05% in 200 mL drinking water daily, for 14 days) significantly promoted angiogenesis and perfusion as well as activation of endothelial AMPK signaling in the ischemic hindlimb. Taken together, caffeine induces mitochondrial fission through cAMP/PKA/AMPK signaling pathway. Mitochondrial fission is an integral process in caffeine-induced endothelial cell migration by altering mitochondrial distribution and energetics.


Subject(s)
Caffeine/therapeutic use , Endothelium/drug effects , Ischemia/drug therapy , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Neovascularization, Physiologic/drug effects , Animals , Cell Movement/drug effects , Hindlimb/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice, Inbred C57BL , Pseudopodia/drug effects , Signal Transduction/drug effects
17.
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
18.
FASEB J ; 35(2): e21287, 2021 02.
Article in English | MEDLINE | ID: mdl-33423335

ABSTRACT

Regulation of sphingolipid metabolism plays a role in cellular homeostasis, and dysregulation of these pathways is involved in cancer progression. Previously, our reports identified ceramide as an anti-metastatic lipid. In the present study, we investigated the biochemical alterations in ceramide-centered metabolism of sphingolipids that were associated with metastatic potential. We established metastasis-prone sublines of SKOV3 ovarian cancer cells using an in vivo selection method. These cells showed decreases in ceramide levels and ceramide synthase (CerS) 2 expression. Moreover, CerS2 downregulation in ovarian cancer cells promoted metastasis in vivo and potentiated cell motility and invasiveness. Moreover, CerS2 knock-in suppressed the formation of lamellipodia required for cell motility in this cell line. In order to define specific roles of ceramide species in cell motility controlled by CerS2, the effect of exogenous long- and very long-chain ceramide species on the formation of lamellipodia was evaluated. Treatment with distinct ceramides increased cellular ceramides and had inhibitory effects on the formation of lamellipodia. Interestingly, blocking the recycling pathway of ceramides by a CerS inhibitor was ineffective in the suppression of exogenous C24:1 -ceramide for the formation of lamellipodia. These results suggested that C24:1 -ceramide, a CerS2 metabolite, predominantly suppresses the formation of lamellipodia without the requirement for deacylation/reacylation. Moreover, knockdown of neutral ceramidase suppressed the formation of lamellipodia concomitant with upregulation of C24:1 -ceramide. Collectively, the CerS2-C24:1 -ceramide axis, which may be countered by neutral ceramidase, is suggested to limit cell motility and metastatic potential. These findings may provide insights that lead to further development of ceramide-based therapy and biomarkers for metastatic ovarian cancer.


Subject(s)
Cell Movement , Ceramides/metabolism , Membrane Proteins/metabolism , Ovarian Neoplasms/metabolism , Pseudopodia/metabolism , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Ceramides/pharmacology , Enzyme Inhibitors/pharmacology , Female , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Ovarian Neoplasms/pathology , Pseudopodia/drug effects , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
19.
Acta Pharmacol Sin ; 42(1): 115-119, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32404981

ABSTRACT

Acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), such as erlotinib, remains a major challenge in the targeted therapy of non-small cell lung cancer (NSCLC). HKB99 is a novel allosteric inhibitor of phosphoglycerate mutase 1 (PGAM1) that preferentially suppresses cell proliferation and induces more apoptosis in acquired erlotinib-resistant HCC827ER cells compared with its parental HCC827 cells. In this study we identified the molecular biomarkers for HKB99 response in erlotinib-resistant HCC827ER cells. We showed that HCC827ER cells displayed enhanced invasive pseudopodia structures as well as downregulated plasminogen activator inhibitor-2 (PAI-2). Meanwhile, PAI-2 knockdown by siPAI-2 candidates decreased the sensitivity of HCC827 parental cells to erlotinib. Moreover, HKB99 (5 µM) preferentially inhibited the invasive pseudopodia formation and increased the level of PAI-2 in HCC827ER cells. Collectively, this study provides new insight into the role of PAI-2 in regulating the sensitivity of erlotinib resistant NSCLC cells to PGAM1 inhibitor. Furthermore, PAI-2 level might be considered as a potential biomarker for predicting the efficacy of the PGAM1 allosteric inhibitor on the erlotinib resistant NSCLC cells.


Subject(s)
Anthracenes/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Enzyme Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/drug therapy , Phosphoglycerate Mutase/antagonists & inhibitors , Sulfonamides/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Erlotinib Hydrochloride/pharmacology , Gene Knockdown Techniques , Humans , Lung Neoplasms/metabolism , Phosphoglycerate Mutase/genetics , Pseudopodia/drug effects , Up-Regulation/drug effects
20.
Int J Mol Sci ; 22(1)2020 Dec 28.
Article in English | MEDLINE | ID: mdl-33379356

ABSTRACT

Hepatocellular carcinoma is one of the most common cancer types worldwide. In cases of advanced-stage disease, sorafenib is considered the treatment of choice. However, resistance to sorafenib remains a major obstacle for effective clinical application. Based on integrated phosphoproteomic and The Cancer Genome Atlas (TCGA) data, we identified a transcription factor, Y-box binding protein-1 (YB-1), with elevated phosphorylation of Ser102 in sorafenib-resistant HuH-7R cells. Phosphoinositide-3-kinase (PI3K) and protein kinase B (AKT) were activated by sorafenib, which, in turn, increased the phosphorylation level of YB-1. In functional analyses, knockdown of YB-1 led to decreased cell migration and invasion in vitro. At the molecular level, inhibition of YB-1 induced suppression of zinc-finger protein SNAI1 (Snail), twist-related protein 1 (Twist1), zinc-finger E-box-binding homeobox 1 (Zeb1), matrix metalloproteinase-2 (MMP-2) and vimentin levels, implying a role of YB-1 in the epithelial-mesenchymal transition (EMT) process in HuH-7R cells. Additionally, YB-1 contributes to morphological alterations resulting from F-actin rearrangement through Cdc42 activation. Mutation analyses revealed that phosphorylation at S102 affects the migratory and invasive potential of HuH-7R cells. Our collective findings suggest that sorafenib promotes YB-1 phosphorylation through effect from the EGFR/PI3K/AKT pathway, leading to significant enhancement of hepatocellular carcinoma (HCC) cell metastasis. Elucidation of the specific mechanisms of action of YB-1 may aid in the development of effective strategies to suppress metastasis and overcome resistance.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Liver Neoplasms/metabolism , Sorafenib/pharmacology , Y-Box-Binding Protein 1/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Invasiveness , Phosphorylation/drug effects , Phosphoserine/metabolism , Prognosis , Pseudopodia/drug effects , Pseudopodia/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Y-Box-Binding Protein 1/genetics , cdc42 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...