Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Am J Pathol ; 191(12): 2203-2218, 2021 12.
Article in English | MEDLINE | ID: mdl-34428425

ABSTRACT

Bladder cancer invasion depends on mammalian target of rapamycin complex 2 (mTORC2) activity, although the downstream mTORC2 effectors that mediate this effect have not been fully defined. One potential downstream effector is the arginine derivative nitric oxide (NO). This study identified a stage-associated increase in the expression of the NO-generating enzymes endothelial NO synthase (eNOS) and inducible NOS (iNOS) in human bladder cancer. Reduction of NOS activity by pharmacologic inhibition or silencing of NOS enzymes reduced cancer cell invasion, with similar effects observed using the NO scavenger cobinamide. By contrast, enhanced invasion was seen with the NO donor Deta-NONOate and an analog of the downstream NO second messenger cGMP. Next, NOS expression was evaluated in invadopodia, which are cellular protrusions that form the invasive tips of cancer cells. Invadopodia were enriched in both iNOS protein and mTORC2 activity, and invadopodia formation was increased by Deta-NONOate and decreased by cobinamide and ablation of mTORC2 activity. Additionally, mTORC2 increased expression of iNOS. Using a zebrafish model, injection of iNOS- or rictor-silenced cells reduced the frequency of bladder cancer cell metastasis in zebrafish. These results indicate that mTORC2 can mediate bladder cancer cell invasion through increased iNOS expression, resulting in increased NO and cGMP production in invadopodia and further propagation of invadopodia formation.


Subject(s)
Mechanistic Target of Rapamycin Complex 2/physiology , Nitric Oxide/metabolism , Podosomes/metabolism , Urinary Bladder Neoplasms/pathology , Animals , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Embryo, Nonmammalian , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Podosomes/genetics , Podosomes/pathology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Zebrafish/embryology
2.
J Exp Clin Cancer Res ; 40(1): 183, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34088337

ABSTRACT

BACKGROUND: Tumor cells are known to release large numbers of exosomes containing active substances that participate in cancer progression. Abnormally expressed long noncoding RNAs (lncRNAs) have been confirmed to regulate multiple processes associated with tumor progression. However, the mechanism by which lncRNAs affect exosome secretion remains unclear. METHODS: The underlying mechanisms of long noncoding RNA LINC00511 (LINC00511) regulation of multivesicular body (MVB) trafficking, exosome secretion, invadopodia formation, and tumor invasion were determined through gene set enrichment analysis (GSEA), immunoblotting, nanoparticle tracking analysis, confocal colocalization analysis, electron microscopy, and invasion experiments. RESULTS: We revealed that the tumorigenesis process is associated with a significant increase in vesicle secretion in hepatocellular carcinoma (HCC). Additionally, LINC00511 was significantly more highly expressed in HCC tissues and is related to vesicle trafficking and MVB distribution. We also found that in addition to the formation of invadopodia in HCC progression, abnormal LINC00511 induces invadopodia formation in HCC cells by regulating the colocalization of vesicle associated membrane protein 7 (VAMP7) and synaptosome associated protein 23 (SNAP23) to induce the invadopodia formation, which are key secretion sites for MVBs and control exosome secretion. Finally, we revealed that LINC0051-induced invadopodia and exosome secretion were involved in tumor progression. CONCLUSIONS: Our experiments revealed novel findings on the relationship between LINC00511 dysregulation in HCC and invadopodia production and exosome secretion. This is a novel mechanism by which LINC00511 regulates invadopodia biogenesis and exosome secretion to further promote cancer progression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , R-SNARE Proteins/genetics , RNA, Long Noncoding/genetics , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Disease Progression , Exosomes/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Podosomes/genetics
3.
J Cell Biol ; 220(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-33904858

ABSTRACT

Podosomes and invadopodia, collectively termed invadosomes, are adhesive and degradative membrane structures formed in many types of cells and are well known for recruiting various proteases. However, another major class of degradative enzymes, deoxyribonuclease (DNase), remains unconfirmed and not studied in invadosomes. Here, using surface-immobilized nuclease sensor (SNS), we demonstrated that invadosomes recruit DNase to their core regions, which degrade extracellular double-stranded DNA. We further identified the DNase as GPI-anchored membrane-bound DNase X. DNase recruitment is ubiquitous and consistent in invadosomes of all tested cell types. DNase activity exhibits within a minute after actin nucleation, functioning concomitantly with protease in podosomes but preceding it in invadopodia. We further showed that macrophages form DNase-active podosome rosettes surrounding bacteria or micropatterned antigen islets, and the podosomes directly degrade bacterial DNA on a surface, exhibiting an apparent immunological function. Overall, this work reports DNase in invadosomes for the first time, suggesting a richer arsenal of degradative enzymes in invadosomes than known before.


Subject(s)
Actins/genetics , DNA/genetics , Deoxyribonucleases/genetics , Cell Physiological Phenomena/genetics , Deoxyribonuclease (Pyrimidine Dimer)/genetics , Extracellular Matrix/genetics , Humans , Podosomes/genetics
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
5.
J Cell Biol ; 219(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32479595

ABSTRACT

Cancer cells break tissue barriers by use of small actin-rich membrane protrusions called invadopodia. Complete invadopodia maturation depends on protrusion outgrowth and the targeted delivery of the matrix metalloproteinase MT1-MMP via endosomal transport by mechanisms that are not known. Here, we show that the ER protein Protrudin orchestrates invadopodia maturation and function. Protrudin formed contact sites with MT1-MMP-positive endosomes that contained the RAB7-binding Kinesin-1 adaptor FYCO1, and depletion of RAB7, FYCO1, or Protrudin inhibited MT1-MMP-dependent extracellular matrix degradation and cancer cell invasion by preventing anterograde translocation and exocytosis of MT1-MMP. Moreover, when endosome translocation or exocytosis was inhibited by depletion of Protrudin or Synaptotagmin VII, respectively, invadopodia were unable to expand and elongate. Conversely, when Protrudin was overexpressed, noncancerous cells developed prominent invadopodia-like protrusions and showed increased matrix degradation and invasion. Thus, Protrudin-mediated ER-endosome contact sites promote cell invasion by facilitating translocation of MT1-MMP-laden endosomes to the plasma membrane, enabling both invadopodia outgrowth and MT1-MMP exocytosis.


Subject(s)
Breast Neoplasms/enzymology , Cell Movement , Endoplasmic Reticulum/enzymology , Endosomes/enzymology , Exocytosis , Matrix Metalloproteinase 14/metabolism , Vesicular Transport Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Endosomes/genetics , Endosomes/pathology , Extracellular Matrix/enzymology , Extracellular Matrix/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 14/genetics , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Invasiveness , Podosomes/enzymology , Podosomes/genetics , Podosomes/pathology , Protein Transport , Signal Transduction , Synaptotagmins/genetics , Synaptotagmins/metabolism , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
6.
J Biol Chem ; 295(31): 10535-10559, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32299913

ABSTRACT

SHC adaptor protein (SHCA) and lipoma-preferred partner (LPP) mediate transforming growth factor ß (TGFß)-induced breast cancer cell migration and invasion. Reduced expression of either protein diminishes breast cancer lung metastasis, but the reason for this effect is unclear. Here, using total internal reflection fluorescence (TIRF) microscopy, we found that TGFß enhanced the assembly and disassembly rates of paxillin-containing adhesions in an SHCA-dependent manner through the phosphorylation of the specific SHCA tyrosine residues Tyr-239, Tyr-240, and Tyr-313. Using a BioID proximity labeling approach, we show that SHCA exists in a complex with a variety of actin cytoskeletal proteins, including paxillin and LPP. Consistent with a functional interaction between SHCA and LPP, TGFß-induced LPP localization to cellular adhesions depended on SHCA. Once localized to the adhesions, LPP was required for TGFß-induced increases in cell migration and adhesion dynamics. Mutations that impaired LPP localization to adhesions (mLIM1) or impeded interactions with the actin cytoskeleton via α-actinin (ΔABD) abrogated migratory responses to TGFß. Live-cell TIRF microscopy revealed that SHCA clustering at the cell membrane preceded LPP recruitment. We therefore hypothesize that, in the presence of TGFß, SHCA promotes the formation of small, dynamic adhesions by acting as a nucleator of focal complex formation. Finally, we defined a previously unknown function for SHCA in the formation of invadopodia, a process that also required LPP. Our results reveal that SHCA controls the formation and function of adhesions and invadopodia, two key cellular structures required for breast cancer metastasis.


Subject(s)
Cell Movement , Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Podosomes/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Animals , Cell Adhesion , Cell Line, Transformed , Cytoskeletal Proteins/genetics , Female , LIM Domain Proteins/genetics , Mice , Paxillin/genetics , Paxillin/metabolism , Podosomes/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Transforming Growth Factor beta
7.
Cancer Lett ; 483: 98-113, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32217106

ABSTRACT

Liver metastases remain a major cause of death from gastrointestinal tract cancers and other malignancies, such as breast and lung carcinomas. Understanding the underlying biology is essential for the design of effective therapies. We previously identified the chemokine CCL7 and its receptor CCR3 as critical mediators of invasion and metastasis in lung and colon carcinoma cells. Here we show that the CCL7/CCR3 axis regulates a late stage in invadopodia genesis namely, the targeting of MMP-9 to the invadopodia complex, thereby promoting invadopodia maturation and collagen degradation. We show that this process could be blocked by overexpression of a dominant negative RhoA in highly invasive cells, while a constitutively active RhoA upregulated invadopodia maturation in CCL7-silenced and poorly invasive and metastatic cells and also enhanced their metastatic potential in vivo, collectively, implicating RhoA activation in signaling downstream of CCL7. Blockade of the ERK or PI3K pathways by chemical inhibitors also inhibited invadopodia formation, but affected the initiation stage of invadopodia genesis. Our data implicate CCL7/CCR3 signaling in invadopodia maturation and suggest that chemokine signaling acts in concert with extracellular matrix-initiated signals to promote invasion and liver metastasis.


Subject(s)
Carcinoma, Lewis Lung/enzymology , Cell Movement , Chemokine CCL7/metabolism , Collagen/metabolism , Colonic Neoplasms/enzymology , Liver Neoplasms/enzymology , Matrix Metalloproteinase 9/metabolism , Podosomes/enzymology , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Chemokine CCL7/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Matrix Metalloproteinase 9/genetics , Mice , Phosphatidylinositol 3-Kinase/metabolism , Podosomes/genetics , Podosomes/pathology , Protein Transport , Proteolysis , Receptors, CCR3/genetics , Receptors, CCR3/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
8.
Biochim Biophys Acta Mol Cell Res ; 1867(7): 118693, 2020 07.
Article in English | MEDLINE | ID: mdl-32198023

ABSTRACT

Talin2 plays an important role in transduction of mechanical signals between extracellular matrix and actin cytoskeleton. Recent studies showed that talin2 is localized to invadopodia and regulates their maturation, subsequently cancer cell invasion and metastasis. However, the molecular mechanism whereby talin2 mediates invadopodium maturation is unknown. Here we show that ablation of talin2 in MDA-MB-231 cells inhibited the secretion of matrix metallopeptidase 9 (MMP9), a proteinase involved in extracellular matrix degradation in invadopodium maturation and metastasis. Furthermore, re-expression of talin2WT in talin2-KO cells rescued MMP9 secretion, but talin2S339C, a mutant with reduced ß-integrin binding, did not, indicating that the talin2-ß-integrin interaction is involved in the MMP9 secretion. Moreover, ablation of talin2 caused an accumulation of enlarged MMP9 vesicles. These vesicles co-localized with enlarged early, late endosomes and autophagosomes, suggesting talin2 controls MMP9 trafficking process. Therefore, these data suggest that talin2 regulates extracellular matrix degradation and invadopodium maturation by mediating MMP9 secretion.


Subject(s)
Matrix Metalloproteinase 9/genetics , Podosomes/genetics , Talin/genetics , Extracellular Matrix/genetics , Gene Expression Regulation/genetics , Humans , Matrix Metalloproteinase 9/biosynthesis , Podosomes/physiology , Protein Binding/genetics , Protein Transport/genetics
9.
J Hematol Oncol ; 13(1): 12, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32033570

ABSTRACT

BACKGROUND: MicroRNA-612 (miR-612) has been proven to suppress EMT, stemness, and tumor metastasis of hepatocellular carcinoma (HCC) via PI3K/AKT2 and Sp1/Nanog signaling. However, its biological roles on HCC progression are far from elucidated. METHODS: We found direct downstream target of miR-612, hadha by RNA immunoprecipitation and sequencing. To explore its biological characteristic, potential molecular mechanism, and clinical relevance in HCC patients, we performed several in-vitro and in-vivo models, as well as human tissue chip. RESULTS: Ectopic expression of miR-612 could partially reverse the level of HADHA, then suppress function of pseudopods, and diminish metastatic and invasive potential of HCC by lipid reprogramming. In detail, miR-612 might reduce invadopodia formation via HADHA-mediated cell membrane cholesterol alteration and accompanied with the inhibition of Wnt/ß-catenin regulated EMT occurrence. Our results showed that the maximum oxygen consumption rates (OCR) of HCCLM3miR-612-OE and HCCLM3hadha-KD cells were decreased nearly by 40% and 60% of their counterparts (p < 0.05). The levels of acetyl CoA were significantly decreased, about 1/3 (p > 0.05) or 1/2 (p < 0.05) of their controls, in exogenous miR-612 or hadha-shRNA transfected HCCLM3 cell lines. Besides, overexpression of hadha cell lines had a high expression level of total cholesterol, especially 27-hydroxycholesterol (p < 0.005). SREBP2 protein expression level as well as its downstream targets, HMGCS1, HMGCR, MVD, SQLE were all deregulated by HADHA. Meanwhile, the ATP levels were reduced to 1/2 and 1/4 in HCCLM3miR-612-OE (p < 0.05) and HCCLM3hadha-KD (p < 0.01) respectively. Moreover, patients with low miR-612 levels and high HADHA levels had a poor prognosis with shorter overall survival. CONCLUSION: miR-612 can suppress the formation of invadopodia, EMT, and HCC metastasis and by HADHA-mediated lipid programming, which may provide a new insight of miR-612 on tumor metastasis and progression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Mitochondrial Trifunctional Protein, alpha Subunit/genetics , Podosomes/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Humans , Lipid Metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Podosomes/pathology
10.
Theranostics ; 10(5): 2358-2373, 2020.
Article in English | MEDLINE | ID: mdl-32104508

ABSTRACT

Invadopodia formation is a key driver of cancer metastasis. The noncanonical IkB-related kinase IKKε has been implicated in cancer metastasis, but its roles in invadopodia formation and colorectal cancer (CRC) metastasis are unclear. Methods: Immunofluorescence, gelatin-degradation assay, wound healing assay and transwell invasion assay were used to determine the influence of IKKε over-expression, knockdown and pharmacological inhibition on invadopodia formation and the migratory and invasive capacity of CRC cells in vitro. Effects of IKKε knockdown or pharmacological inhibition on CRC metastasis were examined in mice. Immunohistochemistry staining was used to detect expression levels of IKKε in CRC patient tissues, and its association with prognosis in CRC patients was also analyzed. Immunoprecipitation, western blotting and in vitro kinase assay were constructed to investigate the molecular mechanisms. Results: IKKε co-localizes with F-actin and the invadopodia marker Tks5 at the gelatin-degrading sites of CRC cells. Genetic over-expression/knockdown or pharmacological inhibition of IKKε altered invadopodia formation and the migratory and invasive capacity of CRC cells in vitro. In vivo, knockdown or pharmacological inhibition of IKKε significantly suppressed metastasis of CRC cells in mice. IKKε knockdown also inhibited invadopodia formation in vivo. Clinical investigation of tumor specimens from 191 patients with CRC revealed that high IKKε expression correlates with metastasis and poor prognosis of CRC. Mechanistically, IKKε directly binds to and phosphorylates kindlin-2 at serine 159; this effect mediates the IKKε-induced invadopodia formation and promotion of CRC metastasis. Conclusions: We identify IKKε as a novel regulator of invadopodia formation and a unique mechanism by which IKKε promotes the metastasis of CRC. Our study suggests that IKKε is a potential target to suppress CRC metastasis.


Subject(s)
Colorectal Neoplasms/pathology , Cytoskeletal Proteins/metabolism , I-kappa B Kinase/metabolism , Muscle Proteins/metabolism , Podosomes/metabolism , Actins/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cytoskeletal Proteins/genetics , Female , Gene Knockdown Techniques , Humans , I-kappa B Kinase/genetics , Male , Mice , Muscle Proteins/genetics , Neoplasm Metastasis , Phosphate-Binding Proteins/metabolism , Phosphorylation , Podosomes/genetics , RNA, Small Interfering/genetics
11.
PLoS One ; 15(1): e0227855, 2020.
Article in English | MEDLINE | ID: mdl-31999741

ABSTRACT

The Src substrate Tks5 helps scaffold matrix-remodeling invadopodia in invasive cancer cells. Focus was directed here on how the five SH3 domains of Tks5 impact that activity. Mutations designed to inhibit protein-protein interactions were created in the individual SH3 domains of Tks5, and the constructs were introduced into the LNCaP prostate carcinoma cell line, a model system with intrinsically low Tks5 expression and which our lab had previously showed the dependence of Src-dependent Tks5 phosphorylation on invadopodia development. In LNCaP cells, acute increases in wild-type Tks5 led to increased gelatin matrix degradation. A similar result was observed when Tks5 was mutated in its 4th or 5th SH3 domains. This was in contrast to the 1st, 2nd, and 3rd SH3 domain mutations of Tks5 where each had a remarkable accentuating effect on gelatin degradation. Conversely, in the invadopodia-competent Src-3T3 model system, mutations in any one of the first three SH3 domains had a dominant negative effect that largely eliminated the presence of invadopodia, inhibited gelatin degradation activity, and redistributed both Src, cortactin, and Tks5 to what are likely endosomal compartments. A hypothesis involving Tks5 conformational states and the regulation of endosomal trafficking is presented as an explanation for these seemingly disparate results.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Carcinoma/genetics , Prostatic Neoplasms/genetics , src-Family Kinases/genetics , Adaptor Proteins, Vesicular Transport/chemistry , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Movement/genetics , Cortactin/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gelatin/genetics , Gelatin/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mutation/genetics , Phosphorylation , Podosomes/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Interaction Domains and Motifs/genetics , src Homology Domains/genetics
12.
J Cell Biol ; 219(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31865373

ABSTRACT

Podosomes are compartmentalized actin-rich adhesions, defined by their ability to locally secrete proteases and remodel extracellular matrix. Matrix remodeling by endothelial podosomes facilitates invasion and thereby vessel formation. However, the mechanisms underlying endothelial podosome formation and function remain unclear. Here, we demonstrate that Septin2, Septin6, and Septin7 are required for maturation of nascent endothelial podosomes into matrix-degrading organelles. We show that podosome development occurs through initial mobilization of the scaffolding protein Tks5 and F-actin accumulation, followed by later recruitment of Septin2. Septin2 localizes around the perimeter of podosomes in close proximity to the basolateral plasma membrane, and phosphoinositide-binding residues of Septin2 are required for podosome function. Combined, our results suggest that the septin cytoskeleton forms a diffusive barrier around nascent podosomes to promote their maturation. Finally, we show that Septin2-mediated regulation of podosomes is critical for endothelial cell invasion associated with angiogenesis. Therefore, targeting of Septin2-mediated podosome formation is a potentially attractive anti-angiogenesis strategy.


Subject(s)
Cell Cycle Proteins/genetics , Neovascularization, Physiologic/genetics , Septins/genetics , Actin Cytoskeleton/genetics , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cell Movement/genetics , Cells, Cultured , Endothelial Cells/metabolism , Extracellular Matrix/genetics , Humans , Morphogenesis/genetics , Podosomes/genetics
13.
Cells ; 8(11)2019 11 05.
Article in English | MEDLINE | ID: mdl-31694343

ABSTRACT

Hepatocellular carcinoma (HCC) represents the fifth most common cancer worldwide and the third cause of cancer-related mortality. Hepatitis C virus (HCV) is the leading cause of chronic hepatitis, which often results in liver fibrosis, cirrhosis, and eventually HCC. HCV is the most common risk factor for HCC in western countries and leads to a more aggressive and invasive disease with poorer patient survival rates. However, the mechanism by which the virus induces the metastatic spread of HCC tumor cells through the regulation of invadopodia, the key features of invasive cancer, is still unknown. Here, the integration of transcriptome with functional kinome screen revealed that HCV infection induced invasion and invadopodia-related gene expression combined with activation of host cell tyrosine kinases, leading to invadopodia formation and maturation and consequent cell invasiveness in vitro and in vivo. The promotion of invadopodia following HCV infection was mediated by the sustained stimulation of epidermal growth factor receptor (EGFR) via the viral NS3/4A protease that inactivates the T-cell protein tyrosine phosphatase (TC-PTP), which inhibits EGFR signaling. Characterization of an invadopodia-associated gene signature in HCV-mediated HCC tumors correlated with the invasiveness of HCC and poor patient prognosis. These findings might lead to new prognostic and therapeutic strategies for virus-mediated invasive cancer.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Hepacivirus/pathogenicity , Hepatitis C/pathology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Neoplasm Invasiveness/pathology , Animals , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , ErbB Receptors/genetics , Gene Expression/genetics , Hepatitis C/virology , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Podosomes/genetics , Podosomes/virology , Prognosis , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Signal Transduction/genetics
14.
Nat Commun ; 10(1): 5171, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729386

ABSTRACT

Basement membrane transmigration during embryonal development, tissue homeostasis and tumor invasion relies on invadosomes, a collective term for invadopodia and podosomes. An adequate structural framework for this process is still missing. Here, we reveal the modular actin nano-architecture that enables podosome protrusion and mechanosensing. The podosome protrusive core contains a central branched actin module encased by a linear actin module, each harboring specific actin interactors and actin isoforms. From the core, two actin modules radiate: ventral filaments bound by vinculin and connected to the plasma membrane and dorsal interpodosomal filaments crosslinked by myosin IIA. On stiff substrates, the actin modules mediate long-range substrate exploration, associated with degradative behavior. On compliant substrates, the vinculin-bound ventral actin filaments shorten, resulting in short-range connectivity and a focally protrusive, non-degradative state. Our findings redefine podosome nanoscale architecture and reveal a paradigm for how actin modularity drives invadosome mechanosensing in cells that breach tissue boundaries.


Subject(s)
Actins/chemistry , Actins/metabolism , Podosomes/metabolism , Actins/genetics , Animals , Cell Adhesion , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Movement , Cells, Cultured , Dendritic Cells/chemistry , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Mechanotransduction, Cellular , Mice , Podosomes/chemistry , Podosomes/genetics
15.
J Cell Mol Med ; 23(12): 8453-8463, 2019 12.
Article in English | MEDLINE | ID: mdl-31638339

ABSTRACT

Epidermal and hepatocyte growth factors can stimulate invasive abilities of melanoma cells, while treatment with combination of their receptors' (EGFR and MET, respectively) inhibitors reduces viability of these cells, as we have previously shown. Proposed therapy has potential; however, used drugs block more than one goal effectively, what raises the question about the real target of analysed inhibitors. For this reason, we analysed direct involvement of these receptors in the invasion of melanoma cells inducing EGFR and MET up- and down-regulations in examined cells. Results were acquired with assays evaluating cell migration and invasion (scratch wound assay, Transwell filter-based method and single-cell tracking). We revealed that cells' motile abilities are increased after EGFR overexpression and decreased following EGFR and MET silencing. This outcome correlates with elevated (EGFR up-regulation) or reduced (EGFR/MET down-regulation) number of formed invadopodia, visualized with immunofluorescence, and their rate of proteolytic abilities, evaluated by fluorescent gelatin degradation assay, and gelatin zymography, compared to control cells. Above-mentioned data indicate that both-EGFR and MET signalling is directly connected with melanoma cells invasion, what establishes these receptors as promising targets for anti-cancer treatment.


Subject(s)
Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Proto-Oncogene Proteins c-met/genetics , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Melanoma/metabolism , Melanoma/pathology , Podosomes/genetics , Podosomes/metabolism , Proto-Oncogene Proteins c-met/metabolism , RNA Interference , Signal Transduction/genetics
16.
Sci Rep ; 9(1): 14363, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31591456

ABSTRACT

Endothelial cell motility has fundamental role in vasculogenesis and angiogenesis during developmental or pathological processes. Tks4 is a scaffold protein known to organize the cytoskeleton of lamellipodia and podosomes, and thus modulating cell motility and invasion. In particular, Tks4 is required for the localization and activity of membrane type 1-matrix metalloproteinase, a key factor for extracellular matrix (ECM) cleavage during cell migration. While its role in transformed cells is well established, little is known about the function of Tks4 under physiological conditions. In this study we examined the impact of Tks4 gene silencing on the functional activity of primary human umbilical vein endothelial cells (HUVEC) and used time-lapse videomicrosopy and quantitative image analysis to characterize cell motility phenotypes in culture. We demonstrate that the absence of Tks4 in endothelial cells leads to impaired ECM cleavage and decreased motility within a 3-dimensional ECM environment. Furthermore, absence of Tks4 also decreases the ability of HUVEC cells to form multicellular sprouts, a key requirement for angiogenesis. To establish the involvement of Tks4 in vascular development in vivo, we show that loss of Tks4 leads sparser vasculature in the fetal chorion in the Tks4-deficient 'nee' mouse strain.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytoskeleton/genetics , Extracellular Matrix/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Cell Movement/genetics , Endothelial Cells/metabolism , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/genetics , Podosomes/genetics , Pseudopodia/genetics , Signal Transduction/genetics
17.
J Cell Sci ; 132(20)2019 10 18.
Article in English | MEDLINE | ID: mdl-31533971

ABSTRACT

The process of tumor cell invasion and metastasis includes assembly of invadopodia, protrusions capable of degrading the extracellular matrix (ECM). The effect of cell cycle progression on invadopodia has not been elucidated. In this study, by using invadopodia and cell cycle fluorescent markers, we show in 2D and 3D cultures, as well as in vivo, that breast carcinoma cells assemble invadopodia and invade into the surrounding ECM preferentially during the G1 phase. The expression (MT1-MMP, also known as MMP14, and cortactin) and localization (Tks5; also known as SH3PXD2A) of invadopodia components are elevated in G1 phase, and cells synchronized in G1 phase exhibit significantly higher ECM degradation compared to the cells synchronized in S phase. The cyclin-dependent kinase inhibitor (CKI) p27kip1 (also known as CDKN1B) localizes to the sites of invadopodia assembly. Overexpression and stable knockdown of p27kip1 lead to contrasting effects on invadopodia turnover and ECM degradation. Taken together, these findings suggest that expression of invadopodia components, as well as invadopodia function, are linked to cell cycle progression, and that invadopodia are controlled by cell cycle regulators. Our results caution that this coordination between invasion and cell cycle must be considered when designing effective chemotherapies.


Subject(s)
Extracellular Matrix/metabolism , G1 Phase , Podosomes/metabolism , Animals , Cell Line , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Extracellular Matrix/genetics , Gene Knockout Techniques , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Mice , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Podosomes/genetics , S Phase
18.
Neoplasia ; 21(10): 1063-1072, 2019 10.
Article in English | MEDLINE | ID: mdl-31521948

ABSTRACT

Ewing sarcoma is a bone tumor most commonly diagnosed in adolescents and young adults. Survival for patients with recurrent or metastatic Ewing sarcoma is dismal and there is a dire need to better understand the mechanisms of cell metastasis specific to this disease. Our recent work demonstrated that microenvironmental stress leads to increased Ewing sarcoma cell invasion through Src activation. Additionally, we have shown that the matricellular protein tenascin C (TNC) promotes metastasis in Ewing sarcoma. A major role of both TNC and Src is mediation of cell-cell and cell-matrix interactions resulting in changes in cell motility, invasion, and adhesion. However, it remains largely unknown, if and how, TNC and Src are linked in these processes. We hypothesized that TNC is a positive regulator of invadopodia formation in Ewing sarcoma through its ability to activate Src. We demonstrate here that both tumor cell endogenous and exogenous TNC can enhance Src activation and invadopodia formation in Ewing sarcoma. We found that microenvironmental stress upregulates TNC expression and this is dampened with application of the Src inhibitor dasatinib, suggesting that TNC expression and Src activation cooperate to promote the invasive phenotype. This work reports the impact of stress-induced TNC expression on enhancing cell invadopodia formation, provides evidence for a feed forward loop between TNC and Src to promote cell metastatic behavior, and highlights a pathway by which microenvironment-driven TNC expression could be therapeutically targeted in Ewing sarcoma.


Subject(s)
Podosomes/metabolism , Sarcoma, Ewing/etiology , Sarcoma, Ewing/metabolism , Tenascin/metabolism , Tumor Microenvironment , src-Family Kinases/metabolism , Cell Line, Tumor , Cells, Cultured , Dasatinib/pharmacology , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Models, Biological , Phosphorylation , Podosomes/genetics , Sarcoma, Ewing/pathology , Stress, Physiological/drug effects , Stress, Physiological/genetics , Tumor Microenvironment/genetics , Wnt Proteins/metabolism
19.
Channels (Austin) ; 13(1): 400-409, 2019 12.
Article in English | MEDLINE | ID: mdl-31510893

ABSTRACT

Voltage gated sodium channels (VGSC) are implicated in cancer cell invasion and metastasis. However, the mechanism by which VGSC increase cell invasiveness and probability of metastasis is still unknown. In this review we outline lesser known functions of VGSC outside of action potential propagation, and the current understanding of the effects of VGSC in cancer. Finally, we discuss possible downstream effects of VGSC activation in cancer cells. After extensive review of the literature, the most likely role of VGSC in cancer is in the invadopodia, the leading edge of metastatic cancer cells. Sodium gradients are used to drive many biological processes in the body, and invadopodia may be similar. The function of the sodium hydrogen exchanger (NHE) and sodium calcium exchanger (NCX) are driven by sodium gradients. Voltage gated calcium channels, activated by membrane depolarization, are also capable of becoming activated in response to VGSC activity. Changes to hydrogen ion exchange or calcium handling have functional consequences for invadopodia and would explain the relationship between VGSC expression and invasiveness of cancer cells.


Subject(s)
Neoplasms/metabolism , Voltage-Gated Sodium Channels/metabolism , Animals , Humans , Neoplasms/genetics , Podosomes/genetics , Podosomes/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Sodium/metabolism , Voltage-Gated Sodium Channels/genetics
20.
EBioMedicine ; 47: 208-220, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31420300

ABSTRACT

BACKGROUND: Metastatic bladder cancer (BLCA) is a lethal disease with an unmet need for study. Transgelin (TAGLN) is an actin-binding protein that affects the dynamics of the actin cytoskeleton indicating its robust potential as a metastasis initiator. Here, we sought to explore the expression pattern of TAGLN and elucidate its specific functioning and mechanisms in BLCA. METHODS: A comprehensive assessment of TAGLN expression in BLCA was performed in three cohorts with a total of 847 patients. The potential effects of TAGLN on BLCA were further determined using clinical genomic analyses that guided the subsequent functional and mechanistic studies. In vitro migration, invasion assays and in vivo metastatic mouse model were performed to explore the biological functions of TAGLN in BLCA cells. Immunofluorescence, western blot and correlation analysis were used to investigate the molecular mechanisms of TAGLN. FINDINGS: TAGLN was highly expressed in BLCA and correlated with advanced prognostic features. TAGLN promoted cell colony formation and cell migration and invasion both in vitro and in vivo by inducing invadopodia formation and epithelial-mesenchymal transition, during which a significant correlation between TAGLN and Slug was observed. The progression-dependent correlation between TGF-ß and TAGLN was analysed at both the cellular and tissue levels, while TGF-ß-mediated migration was abolished by the suppression of TAGLN. INTERPRETATION: Overall, TAGLN is a promising novel prognosis biomarker of BLCA, and its metastatic mechanisms indicate that TAGLN may represent a novel target agent that can be utilized for the clinical management of invasive and metastatic BLCA. FUND: This work was supported by the National Natural Science Foundation of China [81772703, 81672546, 81602253]; the Natural Science Foundation of Beijing [71772219, 7152146]. and Innovative Fund for Doctoral Students of Peking University Health Science Center (BUM2018BSS002). Funders had no role in the design of the study, data collection, data analysis, interpretation, or the writing of this report.


Subject(s)
Epithelial-Mesenchymal Transition , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Podosomes/metabolism , Transforming Growth Factor beta/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Podosomes/genetics , Podosomes/pathology , Prognosis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/mortality , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...