Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
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.
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
4.
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
5.
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
6.
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
7.
J Cell Sci ; 131(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29361525

ABSTRACT

The neuroepithelial cell transforming gene 1A (Net1A, an isoform of Net1) is a RhoA subfamily guanine nucleotide exchange factor (GEF) that localizes to the nucleus in the absence of stimulation, preventing it from activating RhoA. Once relocalized in the cytosol, Net1A stimulates cell motility and extracellular matrix invasion. In the present work, we investigated mechanisms responsible for the cytosolic relocalization of Net1A. We demonstrate that inhibition of MAPK pathways blocks Net1A relocalization, with cells being most sensitive to JNK pathway inhibition. Moreover, activation of the JNK or p38 MAPK family pathway is sufficient to elicit Net1A cytosolic localization. Net1A relocalization stimulated by EGF or JNK activation requires nuclear export mediated by CRM1. JNK1 (also known as MAPK8) phosphorylates Net1A on serine 52, and alanine substitution at this site prevents Net1A relocalization caused by EGF or JNK activation. Glutamic acid substitution at this site is sufficient for Net1A relocalization and results in elevated RhoA signaling to stimulate myosin light chain 2 (MLC2, also known as MYL2) phosphorylation and F-actin accumulation. Net1A S52E expression stimulates cell motility, enables Matrigel invasion and promotes invadopodia formation. These data highlight a novel mechanism for controlling the subcellular localization of Net1A to regulate RhoA activation, cell motility, and invasion.


Subject(s)
Cell Movement , Karyopherins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stress, Physiological , Cell Movement/drug effects , Cytosol/drug effects , Cytosol/metabolism , Epidermal Growth Factor/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Glutamic Acid/metabolism , Humans , MCF-7 Cells , Mutation/genetics , Phosphorylation/drug effects , Phosphoserine/metabolism , Podosomes/drug effects , Podosomes/metabolism , Protein Transport/drug effects , Signal Transduction , Stress, Physiological/drug effects , Tumor Necrosis Factor-alpha/pharmacology , rhoA GTP-Binding Protein/metabolism , Exportin 1 Protein
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Eur J Immunol ; 46(3): 677-88, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26631603

ABSTRACT

Macrophage migration to the focus of infection is a hallmark of the innate immune response. Macrophage spreading, adhesion, and migration through the extracellular matrix require dynamic remodeling of the actin cytoskeleton associated to integrin clustering in podosomes and focal adhesions. Here, we show that prostaglandin E2 (PGE2 ), the main prostaglandin produced by macrophages during inflammation, promote the distinctive dose-dependent formation of podosomes or focal adhesions in macrophages. Low concentrations of PGE2 increased p110γ PI3K expression, phosphorylation of actin-related protein 2, and formation of podosomes, which enhanced macrophage migration in response to chemokines. However, high doses of PGE2 increased phosphorylation of paxillin and focal adhesion kinase, the expression of serine/threonine protein kinase 1, and promoted focal adhesion formation and macrophage adhesion, reducing macrophage chemotaxis. In summary, we describe the dual role of PGE2 as a promoter of macrophage chemotaxis and adhesion, proposing a new model of macrophage migration to the inflammatory focus in the presence of a gradient of PGE2 .


Subject(s)
Cell Adhesion/drug effects , Cell Movement/drug effects , Dinoprostone/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/physiology , Actin-Related Protein 2/genetics , Actin-Related Protein 2/metabolism , Animals , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/ultrastructure , Mice , Paxillin/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Podosomes/drug effects , Protein Kinases/genetics , Signal Transduction/drug effects
14.
Cell Microbiol ; 18(3): 413-23, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26348250

ABSTRACT

Plasmodium falciparum (P. falciparum)-induced effects on the phenotype of human dendritic cells (DC) could contribute to poor induction of long-lasting protective immunity against malaria. DC ability to present antigens to naïve T cells, thus initiating adaptive immune responses depends on complex switches in chemokine receptors, production of soluble mediators and expression of molecules enabling antigen-presentation and maturation. To examine the cellular basis of these processes in the context of malaria, we performed detailed analysis of early events following exposure of human monocyte-derived DC to natural hemozoin (nHZ) and the synthetic analog of its heme core, ß-hematin. DC exposed to either molecule produced high levels of the inflammatory chemokine MCP-1, showed continuous high expression of the inflammatory chemokine receptor CCR5, no upregulation of the lymphoid homing receptor CCR7 and no cytoskeletal actin redistribution with loss of podosomes. DC partially matured as indicated by increased expression of major histocompatibility complex (MHC) class II and CD86 following nHZ and ß-hematin exposure, however there was a lack in expression of the maturation marker CD83 following nHZ but not ß-hematin exposure. Overall our data demonstrate that exposure to nHZ partially impairs the capacity of DC to mature, an effect in part differential to ß-hematin.


Subject(s)
Dendritic Cells/physiology , Hemeproteins/physiology , Host-Parasite Interactions/physiology , Malaria, Falciparum/metabolism , Antigens, CD/metabolism , B7-2 Antigen/metabolism , Chemokine CCL2/metabolism , Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Hemeproteins/pharmacology , Humans , Immunoglobulins/metabolism , Lipopolysaccharides/pharmacology , Malaria, Falciparum/parasitology , Membrane Glycoproteins/metabolism , Podosomes/drug effects , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , CD83 Antigen
15.
Exp Cell Res ; 349(1): 68-76, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27693494

ABSTRACT

Tumor metastasis is the main reason of death for hepatocellular carcinoma (HCC) patients. Cell migration and invasion are two prerequisites for tumor metastasis, in which TRPM7 and MMPs play an important role. In our study, we found that bradykinin (BK) could upregulate the expression of TRPM7 and dynamically regulate the phosphorylation of non-muscle myosin IIA heavy chain (NMHC-IIA) Ser-1943 in HepG2 cells. The influx of Ca2+ via TRPM7 was necessary for elevating the activity of m-calpain and µ-calpain. Additionally, we observed that BK stimulated HepG2 cells to secrete more MMP2 but not MMP9. Src was critical in the process of MMP2 secretion and invadopodia formation. The heat map showed that BDKRB2, TRPM7 and MMP2 had higher overexpression proportions in 25 HCC cell lines. Some clinical specimens of HCC also indicated that BDKRB2 and MMP2 were overexpressed. In conclusion, BK promoted migration and invasion of HCC cells through TRPM7 and MMP2.


Subject(s)
Bradykinin/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , Calcium/metabolism , Calpain/metabolism , Carcinoma, Hepatocellular/enzymology , Exocytosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , Phosphoserine/metabolism , Podosomes/drug effects , Podosomes/metabolism , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Bradykinin B2/metabolism , Signal Transduction/drug effects , TRPM Cation Channels/genetics , Up-Regulation/drug effects , src-Family Kinases/metabolism
16.
Am J Physiol Endocrinol Metab ; 310(5): E355-66, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26670486

ABSTRACT

Advanced glycation end products (AGEs) disturb bone remodeling during aging, and this process is accelerated in diabetes. However, their role in modulation of osteoclast-induced bone resorption is controversial, with some studies indicating that AGEs enhance bone resorption and others showing the opposite effect. We determined whether AGEs present at different stages of osteoclast differentiation affect bone resorption differently. Based on increased levels of tartrate-resistant acid phosphatase (TRAP) and cathepsin K (CTSK), we identified day 4 of induction as the dividing time of cell fusion stage and mature stage in RAW264.7 cell-derived osteoclast-like cells (OCLs). AGE-modified BSA (50-400 µg/ml) or control BSA (100 µg/ml) was then added at the beginning of each stage. Results showed that the presence of AGEs at the cell fusion stage reduced pit numbers, resorption area, and CTSK expression. Moreover, expression of receptor activator of nuclear factor-κB (RANK) as well as the number of TRAP-positive cells, nuclei per OCL, actin rings, and podosomes also decreased. However, the presence of AGEs at the mature stage enlarged the resorption area markedly and increased pit numbers slightly. Intriguingly, only the number of nuclei per OCL and podosomes increased. These data indicate that AGEs biphasically modulate bone resorption activity of OCLs in a differentiation stage-dependent manner. AGEs at the cell fusion stage reduce bone resorption dramatically, mainly via suppression of RANK expression in osteoclast precursors, whereas AGEs at the mature stage enhance bone resorption slightly, most likely by increasing the number of podosomes in mature OCLs.


Subject(s)
Bone Remodeling/drug effects , Bone Resorption/metabolism , Cell Differentiation/drug effects , Glycation End Products, Advanced/pharmacology , Osteoclasts/drug effects , Acid Phosphatase/metabolism , Actins/drug effects , Actins/metabolism , Animals , Blotting, Western , Cathepsin K/metabolism , Cell Line , Cell Nucleus/drug effects , Immunohistochemistry , Isoenzymes/metabolism , Mice , Osteoclasts/cytology , Osteoclasts/metabolism , Podosomes/drug effects , Podosomes/metabolism , Real-Time Polymerase Chain Reaction , Receptor Activator of Nuclear Factor-kappa B/drug effects , Receptor Activator of Nuclear Factor-kappa B/metabolism , Tartrate-Resistant Acid Phosphatase
17.
Biochem Biophys Res Commun ; 468(1-2): 240-7, 2015.
Article in English | MEDLINE | ID: mdl-26518652

ABSTRACT

Temozolomide (TMZ) is one of the few effective anticancer agents against gliomas. However, acquisition of TMZ resistance or adaptation by gliomas is currently a crucial problem, especially increased invasiveness which is critical for the determination of clinical prognosis. This study investigated the molecular regulatory mechanisms of TMZ resistance in gliomas involved in invasiveness, particularly invadopodia formation, a molecular complex formed at the invasive front to cause extracellular matrix degradation during cellular local invasion. The TMZ-resistant clone of the U343 MG human glioma cell line (U343-R cells) was established. U343-R cells demonstrated higher invadopodia formation compared with U343 cells without TMZ resistance (U343-Con cells). Immunoblot analysis of DNA damage-related mitogen-activated protein kinase signals found increased phosphorylation of c-Jun terminal kinase (JNK) and higher activation of its downstream signaling in U343-R cells compared with U343-Con cells. Treatment of U343-R cells with specific inhibitors of JNK or siRNA targeting JNK suppressed up-regulation of invadopodia formation. In addition, paxillin, one of the known JNK effectors which is phosphorylated and affects cell migration, was phosphorylated at serine 178 in JNK activity-dependent manner. Expression of paxillin with mutation of the serine 178 phosphorylation site in U343-R cells blocked invadopodia formation. The present findings suggest that increased formation of invadopodia in U343-R cells is mediated by hyperactivation of JNK-paxillin signaling, and both JNK and paxillin might become targets of novel therapies against TMZ-resistant gliomas.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioma/drug therapy , JNK Mitogen-Activated Protein Kinases/metabolism , Paxillin/metabolism , Podosomes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Dacarbazine/pharmacology , Glioma/metabolism , Glioma/pathology , Humans , Phosphorylation/drug effects , Podosomes/metabolism , Podosomes/pathology , Signal Transduction/drug effects , Temozolomide
18.
J Pharmacol Sci ; 128(2): 78-82, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25986486

ABSTRACT

The role of myosin light chain kinase (MLCK) in inducing podosomes was examined by confocal and electron microscopy. Removal of myosin from the actin core of podosomes using blebbistatin, a myosin inhibitor, resulted in the formation of smaller podosomes. Downregulation of MLCK by the transfection of MLCK small interfering RNA (siRNA) led to the failure of podosome formation. However, ML-7, an inhibitor of the kinase activity of MLCK, failed to inhibit podosome formation. Based on our previous report (Thatcher et al. J.Pharm.Sci. 116 116-127, 2011), we outlined the important role of the actin-binding activity of MLCK in producing smaller podosomes.


Subject(s)
Myosin-Light-Chain Kinase/physiology , Phorbol 12,13-Dibutyrate/pharmacology , Podosomes/drug effects , Podosomes/ultrastructure , Actins/metabolism , Animals , Azepines/pharmacology , Cells, Cultured , Down-Regulation , Microscopy, Immunoelectron , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Naphthalenes/pharmacology , Podosomes/genetics , Protein Binding , RNA, Small Interfering , Rats
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL