Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Virus Res ; 284: 197991, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32360866

ABSTRACT

Macrophages have been identified as key players within the tumor microenvironment, with classically (M1) and alternatively (M2) activated macrophages exhibiting anti-tumoral and pro-tumoral functions, respectively. The goal of this study was to determine the response of macrophage populations to infection with oncolytic vesicular stomatitis virus (VSV). THP-1 monocytes were differentiated into various macrophage subsets and infected with wild-type (rwt virus) or matrix (M) protein mutant (rM51R-M virus) strains of VSV. Monocytes and M2 macrophages were susceptible to infection and killing by both rwt and rM51R-M viruses. rM51R-M virus also increased expression of the M1 markers p-STAT1, CD80, and TNF-α in pro-tumoral M2 macrophages, suggesting reprogramming towards an M1-like pro-inflammatory state. Meanwhile, rwt virus was more effective than rM51R-M virus at replicating in M2 macrophages and at inhibiting the development of invasive podosome structures. This was all in contrast to anti-tumoral M1 macrophages, which remained resistant to VSV-induced cytopathic effects. These results indicate that macrophage populations are differentially susceptible to VSV and that rwt and rM51R-M viruses may modulate the tumor-promoting phenotype of M2 macrophages by distinct mechanisms.


Subject(s)
Cell Differentiation/immunology , Macrophages/classification , Macrophages/virology , Oncolytic Viruses/immunology , Vesiculovirus/immunology , Humans , Macrophages/immunology , Macrophages/pathology , Oncolytic Viruses/pathogenicity , Podosomes/virology , THP-1 Cells , Vesiculovirus/pathogenicity
2.
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
3.
Mol Cancer ; 14: 6, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25608686

ABSTRACT

BACKGROUND: While angiogenesis inhibitors represent a viable cancer therapy, there is preclinical and clinical data to suggest that many tumors develop resistance to such treatments. Moreover, previous studies have revealed a complex association between autophagy and angiogenesis, and their collective influence on tumorigenesis. Autophagy has been implicated in cytoprotection and tumor promotion, and as such may represent an alternative way of targeting apoptosis-resistant cancer cells. This study explored the anti-cancer agent and boswellic acid analog BA145 as an inducer of autophagy and angiogenesis-mediated cytoprotection of tumor cells. METHODS: Flow cytometry, western blotting, and confocal microscopy were used to investigate the role of BA145 mediated autophagy. ELISA, microvessel sprouting, capillary structure formation, aortic ring and wound healing assays were performed to determine the relationship between BA145 triggered autophagy and angiogenesis. Flow cytometery, western blotting, and microscopy were employed to examine the mechanism of BA145 induced cell death and apoptosis. Live imaging and tumor volume analysis were carried out to evaluate the effect of BA145 triggered autophagy on mouse tumor xenografts. RESULTS: BA145 induced autophagy in PC-3 cancer cells and HUVECs significantly impeded its negative regulation on cell proliferation, migration, invasion and tube formation. These effects of BA145 induced autophagy were observed under both normoxic and hypoxic conditions. However, inhibition of autophagy using either pharmacological inhibitors or RNA interference enhanced the BA145 mediated death of these cells. Similar observations were noticed with sunitinib, the anti-angiogenic properties of which were significantly enhanced during combination treatments with autophagy inhibitors. In mouse tumor xenografts, co-treatment with chloroquinone and BA145 led to a considerable reduction in tumor burden and angiogenesis compared to BA145 alone. CONCLUSION: These studies reveal the essential role of BA145 triggered autophagy in the regulation of angiogenesis and cytoprotection. It also suggests that the combination of the autophagy inhibitors with chemotherapy or anti-angiogenic agents may be an effective therapeutic approach against cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Triterpenes/chemistry , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Indoles/pharmacology , Pyrroles/pharmacology , Sunitinib
4.
Prostate ; 74(2): 134-48, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24174371

ABSTRACT

BACKGROUND: The Src tyrosine kinase substrate and adaptor protein Tks5 had previously been implicated in the invasive phenotype of normal and transformed cell types via regulation of cytoskeletal structures called podosomes/invadopodia. The role of Src-Tks5 signaling in invasive prostate cancer, however, had not been previously evaluated. METHODS: We measured the relative expression of Tks5 in normal (n = 20) and cancerous (n = 184, from 92 patients) prostate tissue specimens by immunohistochemistry using a commercially available tumor microarray. We also manipulated the expression and activity of wild-type and mutant Src and Tks5 constructs in the LNCaP and PC-3 prostate cancer cell lines in order to ascertain the role of Src-Tks5 signaling in invadopodia development, matrix-remodeling activity, motility, and invasion. RESULTS: Our studies demonstrated that Src was activated and Tks5 upregulated in high Gleason score prostate tumor specimens and in invasive prostate cancer cell lines. Remarkably, overexpression of Tks5 in LNCaP cells was sufficient to induce invadopodia formation and associated matrix degradation. This Tks5-dependent increase in invasive behavior further depended on Src tyrosine kinase activity and the phosphorylation of Tks5 at tyrosine residues 557 and 619. In PC-3 cells we demonstrated that Tks5 phosphorylation at these sites was necessary and sufficient for invadopodia-associated matrix degradation and invasion. CONCLUSIONS: Our results suggest a general role for Src-Tks5 signaling in prostate tumor progression and the utility of Tks5 as a marker protein for the staging of this disease.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Adenocarcinoma/pathology , Cell Movement/physiology , Cytoskeleton/physiology , Prostatic Neoplasms/pathology , src-Family Kinases/physiology , Adenocarcinoma/physiopathology , Biomarkers, Tumor/physiology , Case-Control Studies , Cell Line, Tumor , Disease Progression , Humans , Immunohistochemistry , Male , Phosphorylation/physiology , Prostatic Neoplasms/physiopathology , Signal Transduction/physiology
5.
Cancer Res ; 72(11): 2833-43, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22467172

ABSTRACT

More than 30% of primary prostate cancers contain a consensus deletion of an approximately 800 kb locus on chromosome 6q15.1. The MAP3K7 gene, which encodes TGF-ß activated kinase-1 (Tak1), is a putative prostate tumor suppressor gene within this region whose precise function remains obscure. In this study, we investigated the role of Tak1 in human and murine prostate cancers. In 50 well-characterized human cancer specimens, we found that Tak1 expression was progressively lost with increasing Gleason grade, both within each cancer and across all cancers. In murine prostate stem cells and Tak1-deficient prostatic epithelial cells, Tak1 loss increased proliferation, migration, and invasion. When prostate stem cells attenuated for Tak1 were engrafted with fetal urogenital mesenchyme, the histopathology of the grafts reflected the natural history of prostate cancer leading from prostatic intraepithelial neoplasia to invasive carcinoma. In the grafts containing Tak1-suppressed prostate stem cells, p38 and c-jun-NH(2)-kinase activity was attenuated and proliferation was increased. Together, our findings functionally validate the proposed tumor suppressor role of Tak1 in prostate cancer.


Subject(s)
MAP Kinase Kinase Kinases/physiology , Prostatic Neoplasms/prevention & control , Tumor Suppressor Proteins/physiology , Animals , Cell Line , Cell Movement , Cell Proliferation , Humans , MAP Kinase Kinase Kinases/analysis , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Mice , Neoplasm Invasiveness , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology
6.
PLoS One ; 7(1): e29761, 2012.
Article in English | MEDLINE | ID: mdl-22238651

ABSTRACT

Invadopodia are membrane protrusions that facilitate matrix degradation and cellular invasion. Although lipids have been implicated in several aspects of invadopodia formation, the contributions of de novo fatty acid synthesis and lipogenesis have not been defined. Inhibition of acetyl-CoA carboxylase 1 (ACC1), the committed step of fatty acid synthesis, reduced invadopodia formation in Src-transformed 3T3 (3T3-Src) cells, and also decreased the ability to degrade gelatin. Inhibition of fatty acid synthesis through AMP-activated kinase (AMPK) activation and ACC phosphorylation also decreased invadopodia incidence. The addition of exogenous 16∶0 and 18∶1 fatty acid, products of de novo fatty acid synthesis, restored invadopodia and gelatin degradation to cells with decreased ACC1 activity. Pharmacological inhibition of ACC also altered the phospholipid profile of 3T3-Src cells, with the majority of changes occurring in the phosphatidylcholine (PC) species. Exogenous supplementation with the most abundant PC species, 34∶1 PC, restored invadopodia incidence, the ability to degrade gelatin and the ability to invade through matrigel to cells deficient in ACC1 activity. On the other hand, 30∶0 PC did not restore invadopodia and 36∶2 PC only restored invadopodia incidence and gelatin degradation, but not cellular invasion through matrigel. Pharmacological inhibition of ACC also reduced the ability of MDA-MB-231 breast, Snb19 glioblastoma, and PC-3 prostate cancer cells to invade through matrigel. Invasion of PC-3 cells through matrigel was also restored by 34∶1 PC supplementation. Collectively, the data elucidate the novel metabolic regulation of invadopodia and the invasive process by de novo fatty acid synthesis and lipogenesis.


Subject(s)
Acetyl-CoA Carboxylase/physiology , Cell Movement/physiology , Cell Surface Extensions/metabolism , Lipogenesis/physiology , Neoplasms/pathology , 3T3 Cells , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Surface Extensions/drug effects , Cell Surface Extensions/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, src/physiology , Humans , Lipogenesis/drug effects , Lipogenesis/genetics , Male , Mice , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/metabolism , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured
7.
Cytoskeleton (Hoboken) ; 68(12): 694-711, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22021214

ABSTRACT

Tks5 is a Src substrate and adaptor protein previously recognized for its regulation of cancer cell invasion through modulation of specialized adhesion structures called podosomes/invadopodia. Here we show for the first time that Tks5 localizes to the podosomes of primary macrophages, and that Tks5 protein levels increase concurrently with podosome deposition during the differentiation of monocytes into macrophages. Similar results are reported for model THP-1 cells, which differentiate into macrophages and form proteolytically active podosomes in response to a PKC signaling agonist (PMA) and with sensitivity to a PKC inhibitor (bisindolylmaleimide). Genetic manipulation of Tks5 expression (silencing and overexpression) in stable THP-1 cell lines does not independently alter this macrophage differentiation process. Nor do these cells lose the ability to focalize F-actin and its accessory proteins into podosome-like structures following PMA treatment. However, Tks5 directly controls podosome-associated gelatin degradation and invasion through collective changes in adhesion, chemotaxis, and the expression/proteolytic activity of MMP9. The Src family kinase-dependent phosphorylation of Tks5 is also implicated in the regulation of THP-1 macrophage invasive behavior. These results therefore define a previously unappreciated function of Tks5 signaling specific to the functional attributes of the macrophage podosome in adhesion, motility, and extracellular matrix-remodeling.


Subject(s)
Adaptor Proteins, Vesicular Transport/blood , Macrophages/cytology , Biomarkers/blood , Cell Line, Tumor , Cell Movement/physiology , Humans , Macrophages/metabolism , Phosphoproteins/metabolism , Signal Transduction
8.
Eur J Cell Biol ; 87(8-9): 555-67, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18417249

ABSTRACT

Podosomes and invadopodia are electron-dense, actin-rich protrusions located on the ventral side of the cellular membrane. They are detected in various types of normal cells, but also in human cancer cells and in Src-transformed fibroblasts. Previously we have shown that the scaffold protein Tks5 (tyrosine kinase substrate 5) co-localizes to podosomes/invadopodia in different human cancer cells and in Src-transformed NIH-3T3 cells. Upon reduced expression of Tks5 podosome formation is decreased, which leads to diminished gelatin degradation in vitro in various human cancer cell lines. It is unclear, however, whether cancer cells need podosomes for tumor growth and metastasis in vivo. To test this idea, we evaluated the ability of Src-transformed NIH-3T3 cells, showing stable reduced expression of Tks5 and podosome formation (Tks5 KD), to form subcutaneous tumors in mice. We demonstrate that decreased expression of Tks5 correlated with reduced tumor growth at this site. In addition, we generated lung metastases from these cells following tail vein injection. The lungs of mice injected i.v. with the Tks5 KD showed smaller-sized metastases, but there was no difference in the number of lesions compared to the controls, indicating that podosomes may not be required for extravasation from the blood stream into the lung parenchyma. Independent of the microenvironment however, the reduced tumor growth correlated with decreased tumor vascularization. Our data potentially implicate a novel role of podosomes as mediators of tumor angiogenesis and support further exploration of how podosome formation and Tks5 expression contribute to tumor progression.


Subject(s)
Microfilament Proteins/physiology , Neoplasms/blood supply , Phosphoproteins/physiology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line, Tumor , Cell Surface Extensions/chemistry , Humans , Immunohistochemistry , Mice , Microfilament Proteins/metabolism , NIH 3T3 Cells , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic , Phosphate-Binding Proteins , Phosphoproteins/metabolism , Transfection , src Homology Domains
9.
Cancer Cell ; 7(2): 155-65, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15710328

ABSTRACT

Tks5/Fish is a scaffolding protein with five SH3 domains and one PX domain. In Src-transformed cells, Tks5/Fish localizes to podosomes, discrete protrusions of the ventral membrane. We generated Src-transformed cells with reduced Tks5/Fish levels. They no longer formed podosomes, did not degrade gelatin, and were poorly invasive. We detected Tks5/Fish expression in podosomes in invasive cancer cells, as well as in human breast cancer and melanoma samples. Tks5/Fish expression was also required for protease-driven matrigel invasion in human cancer cells. Finally, coexpression of Tks5/Fish and Src in epithelial cells resulted in the appearance of podosomes. Thus, Tks5/Fish appears to be required for podosome formation, for degradation of the extracellular matrix, and for invasion of some cancer cells.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Neoplasms/metabolism , Peptide Hydrolases/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Chickens , Extracellular Matrix/metabolism , Humans , Melanoma/metabolism , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Neoplasm Invasiveness , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , src Homology Domains , src-Family Kinases/metabolism
10.
J Biol Chem ; 278(19): 16844-51, 2003 May 09.
Article in English | MEDLINE | ID: mdl-12615925

ABSTRACT

Fish is a scaffolding protein and Src substrate. It contains an amino-terminal Phox homology (PX) domain and five Src homology 3 (SH3) domains, as well as multiple motifs for binding both SH2 and SH3 domain-containing proteins. We have determined that the PX domain of Fish binds 3-phosphorylated phosphatidylinositols (including phosphatidylinositol 3-phosphate and phosphatidylinositol 3,4-bisphosphate). Consistent with this, a fusion protein of green fluorescent protein and the Fish PX domain localized to punctate structures similar to endosomes in normal fibroblasts. However, the full-length Fish protein was largely cytoplasmic, suggesting that its PX domain may not be able to make intermolecular interactions in unstimulated cells. In Src-transformed cells, we observed a dramatic re-localization of some Fish molecules to actin-rich structures called podosomes; the PX domain was both necessary and sufficient to effect this translocation. We used a phage display screen with the fifth SH3 domain of Fish and isolated ADAM19 as a binding partner. Subsequent analyses in mammalian cells demonstrated that Fish interacts with several members of the ADAMs family, including ADAMs 12, 15, and 19. In Src-transformed cells, ADAM12 co-localized with Fish in podosomes. Because members of the ADAMs family have been implicated in growth factor processing, as well as cell adhesion and motility, Fish could be acting as an adaptor molecule that allows Src to impinge on these processes.


Subject(s)
Genes, src/genetics , Metalloendopeptidases/metabolism , Phosphoproteins/metabolism , 3T3 Cells , Animals , COS Cells , Cell Line, Transformed , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/genetics , Mice , Phosphate-Binding Proteins , Phosphoproteins/genetics , Protein Binding , Signal Transduction , src Homology Domains
12.
Plant Physiol ; 130(2): 675-87, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12376635

ABSTRACT

A vacuole membrane-associated calcium-binding protein with an apparent mass of 45 kD was purified from celery (Apium graveolens). This protein, VCaB45, is enriched in highly vacuolate tissues and is located within the lumen of vacuoles. Antigenically related proteins are present in many dicotyledonous plants. VCaB45 contains significant amino acid identity with the dehydrin family signature motif, is antigenically related to dehydrins, and has a variety of biochemical properties similar to dehydrins. VCaB45 migrates anomalously in sodium dodecyl sulfate-polyacrylamide gel electrophoresis having an apparent molecular mass of 45 kD. The true mass as determined by matrix-assisted laser-desorption ionization time of flight was 16.45 kD. VCaB45 has two characteristic dissociation constants for calcium of 0.22 +/- 0.142 mM and 0.64 +/- 0.08 mM, and has an estimated 24.7 +/- 11.7 calcium-binding sites per protein. The calcium-binding properties of VCaB45 are modulated by phosphorylation; the phosphorylated protein binds up to 100-fold more calcium than the dephosphorylated protein. VCaB45 is an "in vitro" substrate of casein kinase II (a ubiquitous eukaryotic kinase), the phosphorylation resulting in a partial activation of calcium-binding activity. The vacuole localization, calcium binding, and phosphorylation of VCaB45 suggest potential functions.


Subject(s)
Calcium-Binding Proteins/metabolism , Plant Proteins/metabolism , Vacuoles/metabolism , Acid Phosphatase/metabolism , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Apium/genetics , Apium/metabolism , Blotting, Western , Calcium/pharmacology , Calcium-Binding Proteins/drug effects , Calcium-Binding Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data , Phosphorylation/drug effects , Phosphotransferases/metabolism , Plant Proteins/genetics , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL