Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Cells ; 13(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38727266

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with high mortality due to early metastatic dissemination and high chemoresistance. All these factors are favored by its extracellular matrix (ECM)-rich microenvironment, which is also highly hypoxic and acidic. Gemcitabine (GEM) is still the first-line therapy in PDAC. However, it is quickly deaminated to its inactive metabolite. Several GEM prodrugs have emerged to improve its cytotoxicity. Here, we analyzed how the acidic/hypoxic tumor microenvironment (TME) affects the response of PDAC cell death and invadopodia-mediated ECM proteolysis to both GEM and its C18 prodrug. METHODS: For this, two PDAC cell lines, PANC-1 and Mia PaCa-2 were adapted to pHe 6.6 or not for 1 month, grown as 3D organotypic cultures and exposed to either GEM or C18 in the presence and absence of acidosis and the hypoxia inducer, deferoxamine. RESULTS: We found that C18 has higher cytotoxic and anti-invadopodia activity than GEM in all culture conditions and especially in acid and hypoxic environments. CONCLUSIONS: We propose C18 as a more effective approach to conventional GEM in developing new therapeutic strategies overcoming PDAC chemoresistance.


Subject(s)
Deoxycytidine , Gemcitabine , Pancreatic Neoplasms , Tumor Microenvironment , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Humans , Tumor Microenvironment/drug effects , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Podosomes/metabolism , Podosomes/drug effects , Drug Resistance, Neoplasm/drug effects , Prodrugs/pharmacology
2.
Molecules ; 27(11)2022 May 28.
Article in English | MEDLINE | ID: mdl-35684426

ABSTRACT

Eucalyptus essential oil and its major constituent eucalyptol are extensively employed in the cosmetic, food, and pharmaceutical industries and their clinical use has recently expanded worldwide as an adjuvant in the treatment of infective and inflammatory diseases. We previously demonstrated that essential oil from Eucalyptus globulus (Labill.) (EO) stimulates in vitro the phagocytic activity of human monocyte-derived macrophages and counteracts the myelotoxicity induced by the chemotherapeutic 5-fluorouracil in immunocompetent rats. Here we characterize some mechanistic aspects underlying the immunostimulatory ability exerted by EO on macrophages. The internalization of fluorescent beads, fluorescent zymosan BioParticles, or apoptotic cancer cells was evaluated by confocal microscopy. Pro-inflammatory cytokine and chemokine release was determined by flow cytometry using the BD cytometric bead array. Receptor involvement in EO-stimulated phagocytosis was assessed using complement- or IgG-opsonized zymosan particles. The localization and expression of podosome components was analyzed by confocal microscopy and western blot. The main results demonstrated that: EO-induced activation of a macrophage is ascribable to its major component eucalyptol, as recently demonstrated for other cells of innate immunity; EO implements pathogen internalization and clearance by stimulating the complement receptor-mediated phagocytosis; EO stimulates podosome formation and increases the expression of podosome components. These results confirm that EO extract is a potent activator of innate cell-mediated immunity and thereby increase the scientific evidence supporting an additional property of this plant extract besides the known antiseptic and anti-inflammatory properties.


Subject(s)
Eucalyptus , Macrophages , Oils, Volatile , Podosomes , Receptors, Complement , Eucalyptol , Eucalyptus/chemistry , Humans , Macrophages/drug effects , Oils, Volatile/pharmacology , Phagocytosis , Podosomes/drug effects , Zymosan
3.
Int J Mol Sci ; 22(21)2021 Nov 07.
Article in English | MEDLINE | ID: mdl-34769479

ABSTRACT

Currently, the etiology of many neuromuscular disorders remains unknown. Many of them are characterized by aberrations in the maturation of the neuromuscular junction (NMJ) postsynaptic machinery. Unfortunately, the molecular factors involved in this process are still largely unknown, which poses a great challenge for identifying potential therapeutic targets. Here, we identified Tks5 as a novel interactor of αdystrobrevin-1, which is a crucial component of the NMJ postsynaptic machinery. Tks5 has been previously shown in cancer cells to be an important regulator of actin-rich structures known as invadosomes. However, a role of this scaffold protein at a synapse has never been studied. We show that Tks5 is crucial for remodeling of the NMJ postsynaptic machinery by regulating the organization of structures similar to the invadosomes, known as synaptic podosomes. Additionally, it is involved in the maintenance of the integrity of acetylcholine receptor (AChR) clusters and regulation of their turnover. Lastly, our data indicate that these Tks5 functions may be mediated by its involvement in recruitment of actin filaments to the postsynaptic machinery. Collectively, we show for the first time that the Tks5 protein is involved in regulation of the postsynaptic machinery.


Subject(s)
Neuromuscular Junction/metabolism , Phosphate-Binding Proteins/physiology , Podosomes/metabolism , Synapses/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neuromuscular Junction/drug effects , Phosphate-Binding Proteins/antagonists & inhibitors , Podosomes/drug effects , Post-Synaptic Density/drug effects , Post-Synaptic Density/metabolism , RNA, Small Interfering/pharmacology , Synapses/drug effects
4.
Cell Rep ; 34(9): 108800, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33657382

ABSTRACT

Cancer cells use actin-based membrane protrusions, invadopodia, to degrade stroma and invade. In serous ovarian cancer (SOC), the endothelin A receptor (ETAR) drives invadopodia by a not fully explored coordinated function of ß-arrestin1 (ß-arr1). Here, we report that ß-arr1 links the integrin-linked kinase (ILK)/ßPIX complex to activate Rac3 GTPase, acting as a central node in the adhesion-based extracellular matrix (ECM) sensing and degradation. Downstream, Rac3 phosphorylates PAK1 and cofilin and promotes invadopodium-dependent ECM proteolysis and invasion. Furthermore, ETAR/ILK/Rac3 signaling supports the communication between cancer and mesothelial cells, favoring SOC cell adhesion and transmigration. In vivo, ambrisentan, an ETAR antagonist, inhibits the adhesion and spreading of tumor cells to intraperitoneal organs, and invadopodium marker expression. As prognostic factors, high EDNRA/ILK expression correlates with poor SOC clinical outcome. These findings provide a framework for the ET-1R/ß-arr1 pathway as an integrator of ILK/Rac3-dependent adhesive and proteolytic signaling to invadopodia, favoring cancer/stroma interactions and metastatic behavior.


Subject(s)
Cell Movement/drug effects , Endothelin-1/pharmacology , Epithelial Cells/enzymology , Ovarian Neoplasms/enzymology , Peritoneum/enzymology , Podosomes/drug effects , Protein Serine-Threonine Kinases/metabolism , Receptor, Endothelin A/metabolism , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Coculture Techniques , Databases, Genetic , Endothelin A Receptor Antagonists/pharmacology , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Peritoneum/pathology , Phenylpropionates/pharmacology , Phosphorylation , Podosomes/enzymology , Podosomes/genetics , Podosomes/pathology , Protein Serine-Threonine Kinases/genetics , Pyridazines/pharmacology , Receptor, Endothelin A/drug effects , Receptor, Endothelin A/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Tumor Microenvironment , Xenograft Model Antitumor Assays , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
6.
Cancer Res ; 81(6): 1472-1485, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33414172

ABSTRACT

Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action. SIGNIFICANCE: An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Endosomes/metabolism , Neoplasm Invasiveness/pathology , Podosomes/drug effects , Protein Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cytoplasmic Dyneins/genetics , Cytoplasmic Dyneins/metabolism , Datasets as Topic , Extracellular Matrix , Female , Gene Expression Profiling , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Male , Melanoma/drug therapy , Melanoma/pathology , Mice , Neoplasm Invasiveness/prevention & control , Podosomes/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Time-Lapse Imaging , Xenograft Model Antitumor Assays
7.
Nanoscale ; 12(17): 9359-9365, 2020 May 07.
Article in English | MEDLINE | ID: mdl-32315013

ABSTRACT

Overactivation and excessive differentiation of osteoclasts (OCs) has been implicated in the course of bone metabolism-related diseases. Although fullerenol nanoparticles (fNPs) have been suggested to inhibit OC differentiation and OC function in our previous work, systemic studies on the effect of fNPs on bone diseases, e.g., osteoporosis (OP), in vivo remain elusive. Herein, it is demonstrated that fNPs significantly suppress the differentiation of OCs that derived from the murine bone marrow monocytes and inhibit the formation of the sealing zone by blocking the formation and patterning of podosomes in OCs spatiotemporally. In vivo, fNPs are supposed to be an efficient inhibitor of the overactivation of OCs in a LPS-induced bone erosion mouse model. The therapeutic effect of fNPs on osteoporosis is also investigated in an ovariectomy-induced osteoporosis rat model. The well-organized trabecular bone, the reduction in the number of TRAP positive cells, the improvement of bone-associated parameters, and the mechanical properties all demonstrate that fNPs, similar to diphosphonates, can be a promising candidate for the effective treatment of osteoporosis.


Subject(s)
Bone Resorption/prevention & control , Fullerenes/therapeutic use , Nanoparticles/therapeutic use , Osteoclasts/drug effects , Osteoporosis/drug therapy , Podosomes/drug effects , Animals , Cancellous Bone/drug effects , Disease Models, Animal , Female , Femur/drug effects , Fullerenes/chemistry , Fullerenes/pharmacology , Mice , Microfilament Proteins/metabolism , Nanoparticles/chemistry , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/drug effects , Osteoporosis/pathology , Osteoporosis/physiopathology , Podosomes/metabolism , Podosomes/pathology , Rats , Tartrate-Resistant Acid Phosphatase/metabolism
8.
J Cell Physiol ; 235(1): 587-598, 2020 01.
Article in English | MEDLINE | ID: mdl-31254281

ABSTRACT

Laminin peptides influence cancer biology. We investigated the role of a laminin-derived peptide C16 regulating invadopodia molecules in human prostate cancer cells (DU145). C16 augmented invadopodia activity of DU145 cells, and stimulated expression Tks4, Tks5, cortactin, and membrane-type matrix metalloproteinase 1. Reactive oxygen species generation is also related to invadopodia formation. This prompted us to address whether C16 would induce reactive oxygen species generation in DU145 cells. Quantitative fluorescence and flow cytometry showed that the peptide C16 increased reactive oxygen species in DU145 cells. Furthermore, significant colocalization between Tks5 and reactive oxygen species was observed in C16-treated cells. Results suggested that the peptide C16 increased Tks5 and reactive oxygen species in prostate cancer cells. The role of C16 increasing Tks and reactive oxygen species are novel findings on invadopodia activity.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Laminin/pharmacology , Podosomes/drug effects , Prostatic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Humans , Laminin/metabolism , Male , Neoplasm Invasiveness/pathology , Prostatic Neoplasms/metabolism , Proteolysis/drug effects
9.
J Cell Physiol ; 235(5): 4655-4666, 2020 05.
Article in English | MEDLINE | ID: mdl-31637713

ABSTRACT

Podosomes and tight junctions (TJs) are subcellular compartments that both exist in endothelial cells and localize at cell surfaces. In contrast to the well-characterized role of TJs in maintaining cerebrovascular integrity, the specific function of endothelial podosomes remains unknown. Intriguingly, we discovered cross-talk between podosomes and TJs in human brain endothelial cells. Tight junction scaffold proteins ZO-1 and ZO-2 localize at podosomes in response to phorbol-12-myristate-13-acetate treatment. We found that both ZO proteins are essential for podosome formation and function. Rather than being derived from new protein synthesis, podosomal ZO-1 and ZO-2 are relocated from a pre-existing pool found at the peripheral plasma membrane with enhanced physical interaction with cortactin, a known protein marker for podosomes. Sequestration of ZO proteins in podosomes weakens tight junction complex formation, leading to increased endothelial cell permeability. This effect can be further attenuated by podosome inhibitor PP2. Altogether, our data revealed a novel cellular function of podosomes, specifically, their ability to negatively regulate tight junction and endothelial barrier integrity, which have been linked to a variety of cerebrovascular diseases.


Subject(s)
Brain/blood supply , Endothelial Cells/metabolism , Podosomes/metabolism , Tight Junctions/metabolism , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-2 Protein/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Humans , Permeability , Podosomes/drug effects , Protein Multimerization , Protein Transport , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Tight Junctions/drug effects , Zonula Occludens-1 Protein/genetics , Zonula Occludens-2 Protein/genetics
10.
Molecules ; 24(14)2019 Jul 18.
Article in English | MEDLINE | ID: mdl-31323836

ABSTRACT

Breast cancer is the most common and the second leading cause of cancer-related deaths in women. It has two distinctive hallmarks: rapid abnormal growth and the ability to invade and metastasize. During metastasis, cancer cells are thought to form actin-rich protrusions, called invadopodia, which degrade the extracellular matrix. Current breast cancer treatments, particularly chemotherapy, comes with adverse effects like immunosuppression, resistance development and secondary tumour formation. Hence, naturally-occurring molecules claimed to be less toxic are being studied as new drug candidates. Ampelopsin E, a natural oligostilbene extracted from Dryobalanops species, has exhibited various pharmacological properties, including anticancer and anti-inflammatory activities. However, there is yet no scientific evidence of the effects of ampelopsin E towards metastasis. Scratch assay, transwell migration and invasion assays, invadopodia and gelatin degradation assays, and ELISA were used to determine the effects of ampelopsin E towards the invasiveness of MDA-MB-231 cells. Strikingly in this study, ampelopsin E was able to halt migration, transmigration and invasion in MDA-MB-231 cells by reducing formation of invadopodia and its degradation capability through significant reduction (p < 0.05) in expression levels of PDGF, MMP2, MMP9 and MMP14. In conclusion, ampelopsin E reduced the invasiveness of MDA-MB-231 cells and was proven to be a potential alternative in treating TNBC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Movement/drug effects , Flavonoids/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dipterocarpaceae/chemistry , Flavonoids/chemistry , Humans , Molecular Structure , Podosomes/drug effects , Triple Negative Breast Neoplasms
11.
Cell Signal ; 61: 39-47, 2019 09.
Article in French | MEDLINE | ID: mdl-31082464

ABSTRACT

Recent study established the role of integrins in keratinocyte growth factor (KGF)-induced oral epithelial adhesion and rete peg elongation. However, how extracellular matrix (ECM) remodeling cooperates with the increased epithelial adhesion during rete peg elongation has yet to be determined. Podosomes are cell-matrix contact structures that combine several abilities, including adhesion and matrix degradation. In the present study, we identified podosome formation at the ventral side of human immortalized oral epithelial cells (HIOECs) upon KGF treatment. Moreover, podosomal components including integrin α6,ß4,α3,ß1 and MMP14 colocalized with the F-actin-cortactin complex and matrix degradation assays demonstrated the ability of the F-actin-cortactin complex to degrade matrix. Inhibition both of integrin subunits ß4 and ß1 with specific blocking antibodies and inhibition of Erk1/2 abrogated the KGF-induced podosome formation. Notably, knockdown of integrin subunits ß4 and ß1 with specific small interfering RNA (siRNA) downregulated the phosphorylation levels of Erk1/2. In contrast, inhibition of both Erk1/2 could upregulate the expression of integrin subunits ß4 and ß1. These results demonstrate that KGF induces podosome formation via integrin-Erk1/2 signaling in HIOECs, suggesting a novel mechanism by which integrins enhance oral epithelial adhesion and rete peg elongation.


Subject(s)
Epithelial Cells/metabolism , Fibroblast Growth Factor 7/pharmacology , Integrin beta1/metabolism , Integrin beta4/metabolism , MAP Kinase Signaling System/drug effects , Mouth Mucosa/cytology , Podosomes/drug effects , Actins/metabolism , Cell Adhesion/drug effects , Cell Line , Cortactin/metabolism , Extracellular Matrix/metabolism , Gene Knockdown Techniques , Humans , Integrin beta1/genetics , Integrin beta4/genetics , Phosphorylation/genetics , Podosomes/metabolism , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Transfection
12.
J Cell Physiol ; 234(11): 20944-20956, 2019 11.
Article in English | MEDLINE | ID: mdl-31020651

ABSTRACT

A series of osteolytic bone diseases are usually related to excessive bone resorption and osteoclast formation. Thus, agents or drugs which can target osteoclast development and attenuate bone loss are potentially considerable in preventing and treating of bone lytic diseases. In recent years, many studies have reported that Notch signaling has substantial impacts on the process of osteoclast differentiation, maturation, and bone destruction. In the present study, we showed that LY411575, a γ-secretase inhibitor, could potently suppress osteoclast differentiation, osteoclast-specific gene expression, and bone resorption via suppressing Notch/HES1/MAPK (ERK and p38)/Akt-mediated NFATc1 induction in vitro. Consistent with in vitro results, LY411575 exhibited protective effects in lipopolysaccharides-induced calvarial bone destruction in vivo. Collectively, these results indicate that LY411575 may have therapeutic potential in the treatment of osteoclast-mediated osteolytic bone diseases.


Subject(s)
Alanine/analogs & derivatives , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Azepines/pharmacology , Enzyme Inhibitors/pharmacology , Osteogenesis/drug effects , Osteolysis/chemically induced , Osteolysis/pathology , Skull/pathology , Actins/metabolism , Alanine/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Resorption/complications , Bone Resorption/genetics , Bone Resorption/pathology , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Fusion , Cell Movement/drug effects , Cell Movement/genetics , Gene Expression Regulation/drug effects , Lipopolysaccharides , Male , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/genetics , Osteolysis/complications , Osteolysis/genetics , Podosomes/drug effects , Podosomes/metabolism , Protective Agents/pharmacology , RANK Ligand/pharmacology , Signal Transduction/drug effects
13.
Sci Rep ; 9(1): 2385, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30787359

ABSTRACT

Angiogenic sprouting can contribute adaptively, or mal-adaptively, to a myriad of conditions including ischemic heart disease and cancer. While the cellular and molecular systems that regulate tip versus stalk endothelial cell (EC) specification during angiogenesis are known, those systems that regulate their distinct actions remain poorly understood. Pre-clinical and clinical findings support sustained adrenergic signaling in promoting angiogenesis, but links between adrenergic signaling and angiogenesis are lacking; importantly, adrenergic agents alter the activation status of the cAMP signaling system. Here, we show that the cAMP effector, PKA, acts in a cell autonomous fashion to constitutively reduce the in vitro and ex vivo angiogenic sprouting capacity of ECs. At a cellular level, we observed that silencing or inhibiting PKA in human ECs increased their invasive capacity, their generation of podosome rosettes and, consequently, their ability to degrade a collagen matrix. While inhibition of either Src-family kinases or of cdc42 reduced these events in control ECs, only cdc42 inhibition, or silencing, significantly impacted them in PKA(Cα)-silenced ECs. Consistent with these findings, cell-based measurements of cdc42 activity revealed that PKA activation inhibits EC cdc42 activity, at least in part, by promoting its interaction with the inhibitory regulator, guanine nucleotide dissociation inhibitor-α (RhoGDIα).


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Endothelial Cells/metabolism , Neovascularization, Pathologic , Neovascularization, Physiologic/drug effects , Podosomes , cdc42 GTP-Binding Protein/physiology , Cell Line , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endothelial Cells/cytology , Endothelial Cells/pathology , Humans , Neovascularization, Physiologic/physiology , Podosomes/drug effects , cdc42 GTP-Binding Protein/antagonists & inhibitors , rho Guanine Nucleotide Dissociation Inhibitor alpha/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/physiology
14.
Antioxid Redox Signal ; 30(14): 1709-1730, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30198307

ABSTRACT

AIMS: Nrf2 (nuclear factor erythroid 2-like 2) is a transcription factor known to modulate blood vessel formation. Various experimental settings, however, attribute to Nrf2 either stimulatory or repressive influence on angiogenesis. Our findings unveil the mechanism of Nrf2-dependent vessel formation, which reaches beyond transactivation of gene expression and reconciles previous discrepancies. RESULTS: We provide evidence that growth differentiation factor 15 (GDF-15)- and stromal cell-derived factor 1 (SDF-1)-induced angiogenesis strongly depends on the presence of Nrf2 protein but does not rely on its transcriptional activity. Instead, Nrf2 serves as a protein restraining Keap1 (Kelch-like ECH-associated protein 1), its known transcriptional repressor. Angiogenic response is abrogated in Nrf2-deficient endothelial cells but not in cells expressing dominant negative form or Keap1-binding fragment of Nrf2. Deficiency of Nrf2 protein available for Keap1 leads to the overabundance of RhoGAP1 (Rho GTPase-activating protein 1), the protein regulating cell division cycle 42 (Cdc42) activity. This impairs podosome assembly and disrupts actin rearrangements, thereby preventing angiogenesis. Effects of Nrf2 deficiency can be rescued by concomitant knockdown of RhoGAP1 or Keap1. Importantly, in the established murine model of Nrf2 deficiency, the N-terminal fragment of Nrf2 containing Keap1 binding domain is preserved. Thus, this model can be used to characterize Nrf2 as a transcription factor, but not as a Keap1-sequestering protein. Innovation and Conclusion: To date, the significance of Nrf2 in cell function has been ascribed solely to the regulation of transcription. We demonstrate that Nrf2 serves as a protein tethering Keap1 to allow podosome assembly and angiogenesis. Moreover, we emphasize that the new Nrf2 function of a Keap1 scavenger implies revisiting the interpretation of some of the previous data on the Nrf2-Keap1 system.


Subject(s)
Endothelial Cells/metabolism , Endothelium/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Podosomes/metabolism , Actins/metabolism , Animals , Cells, Cultured , Cellular Senescence , Chemokine CXCL12 , Endothelial Cells/drug effects , Growth Differentiation Factor 15/pharmacology , High-Throughput Nucleotide Sequencing , Kelch-Like ECH-Associated Protein 1/genetics , Mice , Mice, Knockout , MicroRNAs , Models, Biological , NF-E2-Related Factor 2/genetics , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Podosomes/drug effects , Podosomes/genetics , Transcription, Genetic
15.
Drug Resist Updat ; 39: 1-17, 2018 07.
Article in English | MEDLINE | ID: mdl-30075834

ABSTRACT

Dissemination of cancer cells from the primary tumor and their spread to distant sites of the body is the leading cause of mortality in metastatic cancer patients. Metastatic cancer cells invade surrounding tissues and blood vessels by forming F-actin-rich protrusions known as invadopodia, which degrade the extracellular matrix and enable invasion of tumor cells through it. Invadopodia have now been observed in vivo, and recent evidence demonstrates direct molecular links between assembly of invadopodia and cancer metastasis in both mouse models and in human patients. While significant progress has been achieved in the last decade in understanding the molecular mechanisms and signaling pathways regulating invadopodia formation and function, the application of this knowledge to development of prognostic and therapeutic approaches for cancer metastasis has not been discussed before. Here, we provide a detailed overview of current prognostic markers and tests for cancer metastasis and discuss their advantages, disadvantages, and their predicted efficiency. Using bioinformatic patient database analysis, we demonstrate, for the first time, a significant correlation between invadopodia-associated genes to breast cancer metastasis, suggesting that invadopodia could be used as both a prognostic marker and as a therapeutic target for blocking cancer metastasis. We include here a novel network interaction map of invadopodia-associated proteins with currently available inhibitors, demonstrating a central role for the recently identified EGFR-Pyk2-Src-Arg-cortactin invadopodial pathway, to which re-purposing of existent inhibitors could be used to block breast cancer metastasis. We then present an updated overview of current cancer-related clinical trials, demonstrating the negligible number of trials focusing on cancer metastasis. We also discuss the difficulties and complexity of performing cancer metastasis clinical trials, and the possible development of anti-metastasis drug resistance when using a prolonged preventive treatment with invadopodia inhibitors. This review presents a new perspective on invadopodia-mediated tumor invasiveness and may lead to the development of novel prognostic and therapeutic approaches for cancer metastasis.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Drug Design , Molecular Targeted Therapy , Podosomes/drug effects , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Podosomes/metabolism , Podosomes/pathology , Signal Transduction/drug effects
16.
PLoS One ; 13(6): e0199128, 2018.
Article in English | MEDLINE | ID: mdl-29924834

ABSTRACT

The fucose salvage pathway is a two-step process in which mammalian cells transform L-fucose into GDP-L-fucose, a universal fucose donor used by fucosyltransferases to modify glycans. Emerging evidence indicates the fucose salvage pathway and the fucosylation of proteins are altered during melanoma progression and metastasis. However the underlying mechanisms are not completely understood. Here, we report that the fucose salvage pathway inhibits invadopodia formation and extracellular matrix degradation by promoting α-1,2 fucosylation. Chemically or genetically increasing the fucose salvage pathway decreases invadopodium numbers and inhibits the proteolytic activity of invadopodia in WM793 melanoma cells. Inhibiting fucosylation by depleting fucokinase abrogates L-fucose-mediated inhibition of invadopodia, suggesting dependence on the fucose salvage pathway. The inhibition of invadopodium formation by L-fucose or ectopically expressed FUK could be rescued by treatment with α-1,2, but not α-1,3/α-1,4 fucosidase, implicating an α-1,2 fucose linkage-dependent anti-metastatic effect. The expression of FUT1, an α-1,2 fucosyltransferase, is remarkably down-regulated during melanoma progression, and the ectopic expression of FUT1 is sufficient to inhibit invadopodium formation and ECM degradation. Our findings indicate that the fucose salvage pathway can inhibit invadopodium formation, and consequently, invasiveness in melanoma via α-1,2 fucosylation. Re-activation of this pathway in melanoma could be useful for preventing melanoma invasion and metastasis.


Subject(s)
Extracellular Matrix/metabolism , Fucose/metabolism , Fucosyltransferases/physiology , Melanoma/metabolism , Neoplasm Proteins/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Podosomes/physiology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Fucose/pharmacology , Fucosyltransferases/deficiency , Fucosyltransferases/genetics , Genetic Vectors/pharmacology , Glycosylation , Humans , Melanoma/physiopathology , Metabolic Networks and Pathways , Neoplasm Invasiveness , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Podosomes/drug effects , Protein Processing, Post-Translational , Recombinant Proteins/pharmacology , Up-Regulation , Galactoside 2-alpha-L-fucosyltransferase
17.
J Biol Chem ; 293(26): 10172-10185, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29752407

ABSTRACT

The formin protein dishevelled-associated activator of morphogenesis 1 (DAAM1) polymerizes straight actin filaments and mediates migration of cancer cells. However, how DAAM1 governs cell haptotaxis in response to collagen remains unexplored in breast cancer cells. We hypothesized that DAAM1 mediates invadopodia extension and cell haptotaxis in response to type IV collagen in association with integrin receptors. Using Boyden chamber membranes coated with type IV collagen, we show here that type IV collagen activates both DAAM1 and Ras homolog family member A (RHOA) and promotes haptotaxis of MDA-MB-231 and MDA-MB-453 breast cancer cells, a process abolished by treatment with the integrin αvß3 inhibitor cyclo(-RGDfK). shRNA-mediated knockdown of DAAM1 or a dominant-negative DAAM1 mutation (N-DAAM1) significantly decreased collagen-induced RHOA activity and the assembly of stress fibers, invadopodia extension, and cell haptotaxis. Immunoprecipitation and pulldown assays revealed that integrin αvß3 is associated with, but only indirectly binds to, the C-terminal DAD domain of DAAM1 in mammalian cells. Blockade of RHOA activation with a specific inhibitor (CCG-1423) or via a dominant-negative RHOA mutation (RHOA-N19) suppressed collagen-induced invadopodia extension and haptotaxis of the MDA-MB-231 and MDA-MB-453 cells. Immunoblotting and immunofluorescence assays indicated high DAAM1 and RHOA expression in invadopodia, which was abolished by cyclo(-RGDfK) treatment or DAAM1 knockdown. These findings have uncovered an integrin αvß3/DAAM1/RHOA signaling pathway for type IV collagen-induced invadopodia extension and haptotaxis in breast cancer cells. Targeting this pathway may be a means for reducing invasiveness and metastasis of breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Chemotaxis/drug effects , Collagen/pharmacology , Integrin alphaVbeta3/metabolism , Podosomes/drug effects , Podosomes/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Microfilament Proteins , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism , rho GTP-Binding Proteins , rhoA GTP-Binding Protein/metabolism
18.
J Cell Physiol ; 233(9): 7497-7513, 2018 09.
Article in English | MEDLINE | ID: mdl-29663380

ABSTRACT

Osteoclasts, bone resorbing cells, derive from monocyte/macrophage cell lineage. Increased osteoclast activity is responsible for bone destruction in diseases such as osteoporosis, periodontitis and rheumatoid arthritis. Transglutaminases (TGs), protein crosslinking enzymes, were recently found involved in osteoclastogenesis in vivo, however their mechanisms of action have remained unknown. In this study, we have investigated the role of TG activity in osteoclastogenesis in vitro using four TG inhibitors, NC9, Z006, T101, and monodansyl cadaverine. Our results showed that all TG inhibitors were capable of blocking the entire osteoclastogenesis process. The most potent of the inhibitors, NC9 when added to cultures at different phases of osteoclastogenesis, inhibited differentiation, migration, and fusion of pre-osteoclasts as well as resorption activity of mature osteoclasts. Further investigation into the mechanisms revealed that NC9 increased RhoA levels and blocked podosome belt formation suggesting that TG activity regulates actin dynamics in pre-osteoclasts. The inhibitory effect of NC9 on osteoclastogenesis as well as podosome belt formation was completely reversed with a Rho-family inhibitor Exoenzyme C3. Microtubule architecture, acetylation, and detyrosination of α-tubulin were not affected. Finally, we demonstrated that macrophages and osteoclasts expressed mRNA of three TGs:TG1, TG2, and Factor XIII-A which were all differentially regulated in these cells during differentiation. Immunofluoresence microscopic analysis showed that all three enzymes co-localized to podosomes in osteoclasts. Taken together, our data suggests that TG activity regulates differentiation, migration and fusion of osteoclasts via affecting actin dynamics and that this may involve contribution from all three TG enzymes.


Subject(s)
Actins/metabolism , Cell Differentiation , Cell Movement , Osteoclasts/cytology , Osteoclasts/metabolism , Transglutaminases/metabolism , Animals , Cell Differentiation/drug effects , Cell Fusion , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Podosomes/drug effects , Podosomes/metabolism , Transglutaminases/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
19.
Proc Natl Acad Sci U S A ; 115(12): 3132-3137, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29439204

ABSTRACT

Aberrant activation of endothelin-1 receptors (ET-1R) elicits pleiotropic effects relevant for tumor progression. The network activated by this receptor might be finely, spatially, and temporarily orchestrated by ß-arrestin1 (ß-arr1)-driven interactome. Here, we identify hMENA, a member of the actin-regulatory protein ENA/VASP family, as an interacting partner of ß-arr1, necessary for invadopodial function downstream of ET-1R in serous ovarian cancer (SOC) progression. ET-1R activation by ET-1 up-regulates expression of hMENA/hMENAΔv6 isoforms through ß-arr1, restricted to mesenchymal-like invasive SOC cells. The interaction of ß-arr1 with hMENA/hMENAΔv6 triggered by ET-1 leads to activation of RhoC and cortactin, recruitment of membrane type 1-matrix metalloprotease, and invadopodia maturation, thereby enhancing cell plasticity, transendothelial migration, and the resulting spread of invasive cells. The treatment with the ET-1R antagonist macitentan impairs the interaction of ß-arr1 with hMENA and inhibits invadopodial maturation and tumor dissemination in SOC orthotopic xenografts. Finally, high ETAR/hMENA/ß-arr1 gene expression signature is associated with a poor prognosis in SOC patients. These data define a pivotal function of hMENA/hMENAΔv6 for ET-1/ß-arr1-induced invadopodial activity and ovarian cancer progression.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Endothelin-1/metabolism , Microfilament Proteins/metabolism , Ovarian Neoplasms/pathology , beta-Arrestin 1/metabolism , Animals , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/mortality , Cytoskeleton/metabolism , Endothelin A Receptor Antagonists/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Microfilament Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Podosomes/drug effects , Podosomes/metabolism , Pyrimidines/pharmacology , Receptor, Endothelin A/metabolism , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays , rhoC GTP-Binding Protein/metabolism
20.
Sci Rep ; 8(1): 524, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323185

ABSTRACT

Podosomes are dynamic actin-based membrane protrusions that are important for extracellular matrix degradation and invasive cell motility. Individual podosomes are often found to organize into large rosette-like structures in some types of cells, such as osteoclasts, endothelial cells, Src-transformed fibroblasts, and certain highly invasive cancer cells. In this study, we show that new podosome rosettes arise through one of two mechanisms; de novo assembly or fission of a pre-existing podosome rosette in Src-transformed fibroblasts. Fission is a more efficient way than de novo assembly to generate new podosome rosettes in these cells. Podosome rosettes undergoing fission possess higher motility and a stronger matrix-degrading capability. Podosome rosette fission may be the result of polarized myosin II-mediated contractility of these structures, which is coordinately regulated by myosin light chain kinase and Rho-associated kinase II. Collectively, this study unveils a previously unknown mechanism-fission for the biogenesis of podosome rosettes.


Subject(s)
Podosomes/metabolism , Actins/metabolism , Amides/pharmacology , Animals , Azepines/pharmacology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/physiology , Humans , Mice , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , NIH 3T3 Cells , Naphthalenes/pharmacology , Podosomes/drug effects , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Rosette Formation , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...