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










Publication year range
1.
Cancer Cell ; 37(3): 354-370.e7, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32183951

ABSTRACT

Immunotherapy has emerged as a powerful new chapter in the fight against cancer. However, it has yet to reach its full potential due in part to the complexity of the cancer immune response. We demonstrate that tumor-targeting EDV nanocells function as an immunotherapeutic by delivering a cytotoxin in conjunction with activation of the immune system. These nanocells polarize M1 macrophages and activate NK cells concurrently producing a Th1 cytokine response resulting in potent antitumor function. Dendritic cell maturation and antigen presentation follows, which generates tumor-specific CD8+ T cells, conferring prolonged tumor remission. The combination of cytotoxin delivery and activation of innate and adaptive antitumor immune responses results in a potent cyto-immunotherapeutic with potential in clinical oncology.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Immunity, Innate/drug effects , Salmonella typhimurium/cytology , Adult , Aged , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line , Dendritic Cells/drug effects , Dendritic Cells/physiology , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , ErbB Receptors/administration & dosage , ErbB Receptors/metabolism , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Immunotherapy/methods , Male , Mice , Mice, Inbred BALB C , Nanostructures/chemistry , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology
2.
Sci Rep ; 8(1): 13103, 2018 08 30.
Article in English | MEDLINE | ID: mdl-30166561

ABSTRACT

Recent advances have emphasized the relevance of studying the extracellular microenvironment given its main contribution to tissue homeostasis and disease. Within this complex scenario, we have studied the extracellular protease ADAMTS1 (a disintegrin and metalloprotease with thrombospondin motif 1), implicated in vascularization and development, with reported anti- and pro-tumorigenic activities. In this work we performed a detailed study of the vasculature and substrates in adult organs of wild type and Adamts1-deficient mice. In addition to the expected alterations of organs like kidney, heart and aorta, we found that the lack of ADAMTS1 differently affects lymphocyte and myeloid populations in the spleen and bone marrow. The study of the substrate versican also revealed its alteration in the absence of the protease. With such premises, we challenged our mice with subcutaneous B16F1 syngeneic tumours and closely evaluated the immune repertoire in the tumours but also in the distant spleen and bone marrow. Our results confirmed a pro-inflammatory landscape in the absence of ADAMTS1, correlating with tumour blockade, supporting its novel role as a modulator of the immune cell response.


Subject(s)
ADAMTS1 Protein/metabolism , Inflammation/immunology , Inflammation/pathology , Neoplasms/immunology , Neoplasms/pathology , ADAMTS1 Protein/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Proliferation , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Neoplasms/blood supply , Neoplasms/genetics , Neovascularization, Pathologic/metabolism , Organ Specificity , Spleen/metabolism , Spleen/pathology , Substrate Specificity , Versicans/metabolism
3.
Cancer Lett ; 381(1): 149-55, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27477900

ABSTRACT

We previously showed that BARD1 is a shuttling protein with pro-apoptotic activity in MCF-7 breast cancer cells. BARD1 is expressed as splice variant isoforms in breast cancer. Here we characterized YFP-tagged BARD1 splice variants (beta, omega, phi, ΔRIN, epsilon) for subcellular localization and apoptotic efficacy. We found that loss of nuclear localization (NLS) or export (NES) sequences influenced cellular distribution. The beta and omega variants (+NLS/-NES) shifted exclusively to the nucleus. In contrast, BARD1-epsilon (-NLS/+NES) was mostly cytoplasmic. Variants that lacked both NLS and NES were evenly distributed. Interestingly, the more nuclear isoforms (omega and beta) were least apoptotic in MCF-7 cells as measured by FACS. The cytoplasmic localization of BARD1 isoforms correlated with increased apoptosis. This relationship held in cells exposed to low dose (5 µM) of cisplatin. At 20 µM cisplatin, the main observation was a protective effect by the omega isoform. Similar analyses of HCC1937 cells revealed less pronounced changes but a significant protective influence by BARD1-epsilon. Thus BARD1 variants differ in localization and apoptotic ability, and their expression profile may aid prediction of drug efficacy in breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Active Transport, Cell Nucleus , Alternative Splicing , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cytoplasm/enzymology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , MCF-7 Cells , Protein Isoforms , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Transfection , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics
4.
Oncotarget ; 7(23): 34507-19, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27120788

ABSTRACT

The matrix metalloprotease ADAMTS1 (A Disintegrin And Metalloprotease with ThromboSpondin repeats 1) has been involved in tumorigenesis although its contributions appeared ambiguous. To understand the multifaceted actions of this protease, it is still required a deeper knowledge of its implication in heterogeneous tumor-stroma interactions. Using a syngeneic B16F1 melanoma model in wild type and ADAMTS1 knockout mice we found distinct stroma versus tumor functions for this protease. Genetic deletion of ADAMTS1 in the host microenvironment resulted in a drastic decrease of tumor growth and metastasis. However, the downregulation of tumor ADAMTS1 did not uncover relevant effects. Reduced tumors in ADAMTS1 KO mice displayed a paradoxical increase in vascular density and vascular-related genes; a detailed characterization revealed an impaired vasculature, along with a minor infiltration of macrophages. In addition, ex-vivo assays supported a chief role for ADAMTS1 in vascular sprouting, and melanoma xenografts showed a relevant induction of its expression in stroma compartments. These findings provide the first genetic evidence that supports the pro-tumorigenic role of stromal ADAMTS1.


Subject(s)
ADAMTS1 Protein/genetics , Melanoma, Experimental/pathology , Melanoma/pathology , Neovascularization, Pathologic/pathology , Uveal Neoplasms/pathology , ADAMTS1 Protein/metabolism , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation , Gene Deletion , HEK293 Cells , Humans , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/genetics , Xenograft Model Antitumor Assays
5.
PLoS One ; 11(3): e0150484, 2016.
Article in English | MEDLINE | ID: mdl-26930278

ABSTRACT

Activation of the wnt signaling pathway is a major cause of colon cancer development. Tankyrase inhibitors (TNKSi) have recently been developed to block the wnt pathway by increasing axin levels to promote degradation of the wnt-regulator ß-catenin. TNKSi bind to the PARP (poly(ADP)ribose polymerase) catalytic region of tankyrases (TNKS), preventing the PARylation of TNKS and axin that normally control axin levels through ubiquitination and degradation. TNKSi treatment of APC-mutant SW480 colorectal cancer cells can induce axin puncta which act as sites for assembly of ß-catenin degradation complexes, however this process is poorly understood. Using this model system, we found that siRNA knockdown of TNKSs 1 and 2 actually blocked the ability of TNKSi drugs to induce axin puncta, revealing that puncta formation requires both the expression and the inactivation of TNKS. Immunoprecipitation assays showed that treatment of cells with TNKSi caused a strong increase in the formation of axin-TNKS complexes, correlating with an increase in insoluble or aggregated forms of TNKS/axin. The efficacy of TNKSi was antagonized by proteasome inhibitors, which stabilized the PARylated form of TNKS1 and reduced TNKSi-mediated assembly of axin-TNKS complexes and puncta. We hypothesise that TNKSi act to stimulate TNKS oligomerization and assembly of the TNKS-axin scaffold that form puncta. These new insights may help in optimising the future application of TNKSi in anticancer drug design.


Subject(s)
Axin Protein/metabolism , Tankyrases/antagonists & inhibitors , beta Catenin/metabolism , Animals , Antineoplastic Agents/pharmacology , Axin Protein/drug effects , Blotting, Western , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , Fluorescent Antibody Technique , HEK293 Cells , Humans , Mice , Tankyrases/drug effects , Tankyrases/metabolism , Wnt Signaling Pathway/drug effects
6.
J Cell Sci ; 128(21): 3933-46, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26403202

ABSTRACT

ß-Catenin transduces the Wnt signaling pathway and its nuclear accumulation leads to gene transactivation and cancer. Rac1 GTPase is known to stimulate ß-catenin-dependent transcription of Wnt target genes and we confirmed this activity. Here we tested the recent hypothesis that Rac1 augments Wnt signaling by enhancing ß-catenin nuclear import; however, we found that silencing/inhibition or up-regulation of Rac1 had no influence on nuclear accumulation of ß-catenin. To better define the role of Rac1, we employed proximity ligation assays (PLA) and discovered that a significant pool of Rac1-ß-catenin protein complexes redistribute from the plasma membrane to the nucleus upon Wnt or Rac1 activation. More importantly, active Rac1 was shown to stimulate the formation of nuclear ß-catenin-lymphoid enhancer factor 1 (LEF-1) complexes. This regulation required Rac1-dependent phosphorylation of ß-catenin at specific serines, which when mutated (S191A and S605A) reduced ß-catenin binding to LEF-1 by up to 50%, as revealed by PLA and immunoprecipitation experiments. We propose that Rac1-mediated phosphorylation of ß-catenin stimulates Wnt-dependent gene transactivation by enhancing ß-catenin-LEF-1 complex assembly, providing new insight into the mechanism of cross-talk between Rac1 and canonical Wnt/ß-catenin signaling.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/metabolism , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/physiology , Animals , Blotting, Western , Cell Line , HCT116 Cells , Humans , Immunoprecipitation , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , NIH 3T3 Cells , Real-Time Polymerase Chain Reaction , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/genetics , rac1 GTP-Binding Protein/genetics
7.
Cell Signal ; 27(9): 1763-71, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26022179

ABSTRACT

BARD1 is a breast cancer tumor suppressor with multiple domains and functions. BARD1 comprises a tandem BRCT domain at the C-terminus, and this sequence has been reported to target BARD1 to distinct subcellular locations such as nuclear DNA breakage sites and the centrosome through binding to regulatory proteins such as HP1 and OLA1, respectively. We now identify the BRCT domain as a binding site for p53. We first confirmed previous reports that endogenous BARD1 binds to p53 by immunoprecipitation assay, and further show that BARD1/p53 complexes locate at mitochondria suggesting a cellular location for p53 regulation of BARD1 apoptotic activity. We used a proximity ligation assay to map three distinct p53 binding sequences in human BARD1, ranging from weak (425-525) and modest (525-567) to strong (551-777 comprising BRCT domains). Deletion of the BRCT sequence caused major defects in the ability of BARD1 to (1) bind p53, (2) localize to the cytoplasm and mitochondria, and (3) induce Bax oligomerization and apoptosis. Our data suggest that BARD1 can move to mitochondria independent of p53, but subsequently associates with p53 to induce Bax clustering in part by decreasing mitochondrial Bcl-2 levels. We therefore identify a role for the BRCT domain in stimulating BARD1 nuclear export and mitochondrial localization, and in assembling mitochondrial BARD1/p53 complexes to regulate specific activities such as apoptotic function.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Cytoplasm/metabolism , Mitochondria/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cytoplasm/genetics , DNA Breaks , Female , Humans , MCF-7 Cells , Mitochondria/genetics , Mitochondria/pathology , Protein Structure, Tertiary , Protein Transport/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Sequence Deletion , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
8.
Oncotarget ; 5(20): 9889-900, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25301724

ABSTRACT

5-fluorouracil (5-FU) is the first line component used in colorectal cancer (CRC) therapy however even in combination with other chemotherapeutic drugs recurrence is common. Mutations of the adenomatous polyposis coli (APC) gene are considered as the initiating step of transformation in familial and sporadic CRCs. We have previously shown that APC regulates the cellular response to DNA replication stress and recently hypothesized that APC mutations might therefore influence 5-FU resistance. To test this, we compared CRC cell lines and show that those expressing truncated APC exhibit a limited response to 5-FU and arrest in G1/S-phase without undergoing lethal damage, unlike cells expressing wild-type APC. In SW480 APC-mutant CRC cells, 5-FU-dependent apoptosis was restored after transient expression of full length APC, indicating a direct link between APC and drug response. Furthermore, we could increase sensitivity of APC truncated cells to 5-FU by inactivating the Chk1 kinase using drug treatment or siRNA-mediated knockdown. Our findings identify mutant APC as a potential tumor biomarker of resistance to 5-FU, and importantly we show that APC-mutant CRC cells can be made more sensitive to 5-FU by use of Chk1 inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , DNA Replication/drug effects , DNA, Neoplasm/biosynthesis , Fluorouracil/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Caco-2 Cells , Checkpoint Kinase 1 , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA, Neoplasm/genetics , Drug Synergism , Fluorouracil/administration & dosage , Genes, APC , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Protein Kinase Inhibitors/administration & dosage
9.
Oncotarget ; 5(12): 4295-304, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24962328

ABSTRACT

Expression of IGFBP2 (Insulin-like Growth Factor Binding Protein 2) has been positively correlated with glioma progression. Although the proteolysis of IGFBP2 has been widely recognized, with consequences as a major modulator of IGFII signaling, the relevance of this post-translational modification has not been well studied in tumors. Using an in vivo proteomic approach by Isotope-Coded Protein Label (ICPL), we identified IGFBP2 as a target of the extracellular protease ADAMTS1 (A Disintegrin And Metalloproteinase with ThromboSpondin motifs 1). Notably, the proteolytic pattern of IGFBP2 was also detected in human glioma culture cells and, more importantly, in all glioma samples evaluated. In addition, high expression of ADAMTS1 correlates with higher levels of cleaved IGFBP2 in glioblastoma multiforme cases. Using gene expression public databases, we confirmed that IGFBP2 is a poor prognosis marker for gliomas, and we also observed an important contribution of ADAMTS1.Finally, we showed the impact of ADAMTS1 on IGFII-mediated IGF1R phosphorylation and cellular migration. Our results support a functional interaction between IGFBP2 and ADAMTS1 and suggest the need to evaluate post-translational modifications of IGFBP2 in glioma, in order to approach new therapies.


Subject(s)
ADAM Proteins/genetics , ADAM Proteins/metabolism , Brain Neoplasms/genetics , Glioma/genetics , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , ADAMTS1 Protein , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Phosphorylation , Proteolysis , Signal Transduction , Transfection
10.
Int J Cancer ; 133(10): 2315-24, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23681936

ABSTRACT

The extracellular protease ADAMTS1 (A disintegrin and metalloprotease with thrombospondin repeats 1) has been described as an anti-angiogenic molecule and its role as a putative tumor protective molecule has also been suggested. Here, we have used a tumor xenograft model to determine the role of ADAMTS1 in tumor growth and angiogenesis. Increasing levels of the protease led to the complete inhibition of tumor growth. In an attempt to elucidate the mechanism of action of this protease, we focused our attention on its proteolytic activity on nidogens, one of the main components of the vascular basement membrane. The increased expression of ADAMTS1 was accompanied by increased proteolysis of nidogen-1 and -2 and their almost complete removal from vascular structures, together with major morphological alterations of tumor blood vessels and a decreased vessel density. The clinical relevance of this work is supported by our observations that ADAMTS1 expression is decreased in breast tumor specimens when compared with healthy tissue. Our studies also reveal that the cleavage of nidogen-1 and -2 is partially inhibited in human tumor samples. Moreover, the deposition of both nidogens surrounding vascular structures is drastically altered, implying a possible reduction in the maintenance of vessel integrity. Our studies reflect the requirement to explore the functional interactions between proteases and specific substrates in cancer biology.


Subject(s)
ADAM Proteins/metabolism , Breast Neoplasms/genetics , Cell Adhesion Molecules/metabolism , Genes, Tumor Suppressor , Membrane Glycoproteins/metabolism , Peptide Hydrolases/metabolism , ADAM Proteins/genetics , ADAMTS1 Protein , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium-Binding Proteins , Cell Adhesion Molecules/genetics , Cell Line , Down-Regulation , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Glycosaminoglycans/physiology , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Proteolysis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Nature ; 465(7299): 813-7, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20535211

ABSTRACT

Down's syndrome (DS) is a genetic disorder caused by full or partial trisomy of human chromosome 21 and presents with many clinical phenotypes including a reduced incidence of solid tumours. Recent work with the Ts65Dn model of DS, which has orthologues of about 50% of the genes on chromosome 21 (Hsa21), has indicated that three copies of the ETS2 (ref. 3) or DS candidate region 1 (DSCR1) genes (a previously known suppressor of angiogenesis) is sufficient to inhibit tumour growth. Here we use the Tc1 transchromosomic mouse model of DS to dissect the contribution of extra copies of genes on Hsa21 to tumour angiogenesis. This mouse expresses roughly 81% of Hsa21 genes but not the human DSCR1 region. We transplanted B16F0 and Lewis lung carcinoma tumour cells into Tc1 mice and showed that growth of these tumours was substantially reduced compared with wild-type littermate controls. Furthermore, tumour angiogenesis was significantly repressed in Tc1 mice. In particular, in vitro and in vivo angiogenic responses to vascular endothelial growth factor (VEGF) were inhibited. Examination of the genes on the segment of Hsa21 in Tc1 mice identified putative anti-angiogenic genes (ADAMTS1and ERG) and novel endothelial cell-specific genes, never previously shown to be involved in angiogenesis (JAM-B and PTTG1IP), that, when overexpressed, are responsible for inhibiting angiogenic responses to VEGF. Three copies of these genes within the stromal compartment reduced tumour angiogenesis, explaining the reduced tumour growth in DS. Furthermore, we expect that, in addition to the candidate genes that we show to be involved in the repression of angiogenesis, the Tc1 mouse model of DS will permit the identification of other endothelium-specific anti-angiogenic targets relevant to a broad spectrum of cancer patients.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Disease Models, Animal , Down Syndrome/genetics , Gene Dosage/genetics , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS1 Protein , Animals , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chromosomes, Mammalian/genetics , Down Syndrome/complications , Down Syndrome/physiopathology , Female , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Intracellular Signaling Peptides and Proteins , Male , Melanoma, Experimental/complications , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Transcription Factors , Transcriptional Regulator ERG , Trisomy/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Cancer Res ; 70(11): 4676-86, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20484033

ABSTRACT

Cancer stem cells have been hypothesized to explain tumor plasticity, including the capability to adopt distinct differentiation commitments. Among the mechanisms of tumor neovascularization, the ability of some malignant cells to mimic an endothelial phenotype has been recognized by a capacity to form matrix-enriched pseudovascular structures. In addition to the expression of genes associated with an endothelial nature, the molecular dynamism of specific microenvironments may also be critical. Here, we report the identification of the extracellular protease ADAMTS1 as a critical molecule for tumor cells to acquire endothelial-like properties. In a fibrosarcoma model, ADAMTS1 increased tumor growth rate in an angiogenesis-independent manner, influencing the tumor cells to display an exclusive endothelial-like gene signature. We documented the relevant expression of ADAMTS1 in aggressive and highly plastic melanoma and Ewing sarcoma cells. Notably, inhibiting ADAMTS1 action compromised the endothelial mimetic attributes observed in this setting. Our findings provide insights into how the tumor microenvironment can elicit endothelial mimicry by tumor cells.


Subject(s)
ADAM Proteins/biosynthesis , Melanoma/enzymology , Melanoma/pathology , Sarcoma, Ewing/enzymology , Sarcoma, Ewing/pathology , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , ADAMTS1 Protein , Animals , Cell Line, Tumor , Endothelial Cells/enzymology , Endothelial Cells/pathology , Fibrosarcoma/enzymology , Fibrosarcoma/pathology , Humans , Mice , Mice, Inbred BALB C , Phenotype , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...