Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 141(1): 178-90, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20371353

ABSTRACT

Our findings that PlGF is a cancer target and anti-PlGF is useful for anticancer treatment have been challenged by Bais et al. Here we take advantage of carcinogen-induced and transgenic tumor models as well as ocular neovascularization to report further evidence in support of our original findings of PlGF as a promising target for anticancer therapies. We present evidence for the efficacy of additional anti-PlGF antibodies and their ability to phenocopy genetic deficiency or silencing of PlGF in cancer and ocular disease but also show that not all anti-PlGF antibodies are effective. We also provide additional evidence for the specificity of our anti-PlGF antibody and experiments to suggest that anti-PlGF treatment will not be effective for all tumors and why. Further, we show that PlGF blockage inhibits vessel abnormalization rather than density in certain tumors while enhancing VEGF-targeted inhibition in ocular disease. Our findings warrant further testing of anti-PlGF therapies.


Subject(s)
Neovascularization, Physiologic/drug effects , Pregnancy Proteins/antagonists & inhibitors , Pregnancy Proteins/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/prevention & control , Choroid/blood supply , Disease Models, Animal , Eye Diseases/pathology , Humans , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Papilloma/blood supply , Papilloma/chemically induced , Papilloma/prevention & control , Placenta Growth Factor , Skin Neoplasms/blood supply , Skin Neoplasms/chemically induced , Skin Neoplasms/prevention & control
2.
Cell ; 136(5): 839-851, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19217150

ABSTRACT

A key function of blood vessels, to supply oxygen, is impaired in tumors because of abnormalities in their endothelial lining. PHD proteins serve as oxygen sensors and may regulate oxygen delivery. We therefore studied the role of endothelial PHD2 in vessel shaping by implanting tumors in PHD2(+/-) mice. Haplodeficiency of PHD2 did not affect tumor vessel density or lumen size, but normalized the endothelial lining and vessel maturation. This resulted in improved tumor perfusion and oxygenation and inhibited tumor cell invasion, intravasation, and metastasis. Haplodeficiency of PHD2 redirected the specification of endothelial tip cells to a more quiescent cell type, lacking filopodia and arrayed in a phalanx formation. This transition relied on HIF-driven upregulation of (soluble) VEGFR-1 and VE-cadherin. Thus, decreased activity of an oxygen sensor in hypoxic conditions prompts endothelial cells to readjust their shape and phenotype to restore oxygen supply. Inhibition of PHD2 may offer alternative therapeutic opportunities for anticancer therapy.


Subject(s)
Blood Vessels/cytology , DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Immediate-Early Proteins/metabolism , Neoplasm Metastasis , Neoplasms/blood supply , Oxygen/metabolism , Animals , Blood Vessels/embryology , Blood Vessels/metabolism , Cell Shape , DNA-Binding Proteins/genetics , Endothelial Cells/cytology , Glycolysis , Heterozygote , Hypoxia/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Immediate-Early Proteins/genetics , Mice , Neoplasms/pathology , Procollagen-Proline Dioxygenase
3.
Semin Cancer Biol ; 60: 214-224, 2020 02.
Article in English | MEDLINE | ID: mdl-31386907

ABSTRACT

Cancer is a multistep disease based on crucial interactions between tumor cells and the microenvironment (extracellular matrix and stroma/immune cells). In fact, during dissemination, tumor cells have to escape from the primary tumor mass, cross the basal membrane, interact with endothelial cells to enter blood vessels (intravasation), survive in the bloodstream, get in contact with endothelial cells again to exit the bloodstream (extravasation) and seed in distant organs. Interactions between tumor and stroma cells are strongly coordinated by microRNAs (miRNAs), small non-coding RNAs able to silence protein coding genes by binding to specific recognition sites, mostly located at the 3' UTR of mature mRNAs. Relevantly, miRNA expression is often altered (overexpression or downregulation) in tumor cells and influenced by stroma cells. At the same time, miRNAs are abundant and essential in stroma cells during tumor cell dissemination and their expression is influenced by tumor cells. In fact, for instance, conditional ablation of Dicer in the endothelium of tumor bearing-mice leads to reduced tumor growth and microvessel density. In this review, we specifically focus on the role of miRNAs in endothelial cells regarding their positive or negative intervention on tumor angiogenesis or lymphoangiogenesis or when tumor cells detach from the tumor mass and intravasate or extravasate in/out of the blood vessels. Examples of pro-angiogenic miRNAs are miR-9 or miR-494, often overexpressed in tumors, which accumulate in tumor cell microvescicles and, therefore, get transferred to endothelial cells where they induce migration and angiogenesis. Differently, miR-200 and miR-128 are often downregulated in tumors and inhibit angiogenesis and lymphoangiogenesis. Instead, miR-126 controls intravasation while miR-146a, miR-214, miR-148b govern extravasation, in a positive or negative manner. Finally, at the end, we summarize opportunities for therapeutic interventions based on miRNAs acting on endothelial cells.


Subject(s)
Cell Communication/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/etiology , Neoplasms/metabolism , Tumor Microenvironment/genetics , Animals , Cell Communication/immunology , Disease Progression , Humans , Neoplasm Staging , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Tumor Microenvironment/immunology
4.
Mol Ther ; 26(8): 2008-2018, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29929788

ABSTRACT

We previously demonstrated that miR-214 is upregulated in malignant melanomas and triple-negative breast tumors and promotes metastatic dissemination by affecting a complex pathway including the anti-metastatic miR-148b. Importantly, tumor dissemination could be reduced by blocking miR-214 function or increasing miR-148b expression or by simultaneous interventions. Based on this evidence, with the intent to explore the role of miR-214 as a target for therapy, we evaluated the capability of new chemically modified anti-miR-214, R97/R98, to inhibit miR-214 coordinated metastatic traits. Relevantly, when melanoma or breast cancer cells were transfected with R97/R98, anti-miR-214 reduced miR-214 expression and impaired transendothelial migration were observed. Noteworthy, when the same cells were injected in the tail vein of mice, cell extravasation and metastatic nodule formation in lungs were strongly reduced. Thus, suggesting that R97/R98 anti-miR-214 oligonucleotides were able to inhibit tumor cell escaping through the endothelium. More importantly, when R97/R98 anti-miR-214 compounds were systemically delivered to mice carrying melanomas or breast or neuroendocrine pancreatic cancers, a reduced number of circulating tumor cells and lung or lymph node metastasis formation were detected. Similar results were also obtained when AAV8-miR-214 sponges were used in neuroendocrine pancreatic tumors. Based on this evidence, we propose miR-214 as a promising target for anti-metastatic therapies.


Subject(s)
Antagomirs/administration & dosage , MicroRNAs/genetics , Neoplasms/drug therapy , Up-Regulation/drug effects , Animals , Antagomirs/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/genetics , Cell Line, Tumor , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Melanoma/drug therapy , Melanoma/genetics , Mice , MicroRNAs/antagonists & inhibitors , Neoplasm Metastasis/drug therapy , Neoplasms/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Transfection , Xenograft Model Antitumor Assays
5.
Biochim Biophys Acta ; 1859(10): 1322-32, 2016 10.
Article in English | MEDLINE | ID: mdl-27344374

ABSTRACT

In mammals the cell-cycle progression through the G1 phase is a tightly regulated process mediated by the transcriptional activation of early genes in response to mitogenic stimuli, whose dysregulation often leads to tumorigenesis. We here report the discovery by RNA-seq of cell-cycle regulated (CCR) long intergenic non-coding RNAs (lincRNAs), potentially involved in the control of the cell-cycle progression. We identified 10 novel lincRNAs expressed in response to serum treatment in mouse embryonic fibroblasts (MEFs) and in BALB/c fibroblasts, comparably to early genes. By loss-of-function experiments we found that lincRNA CCR492 is required for G1/S progression, localizes in the cell cytoplasm and contains 4 let-7 microRNA recognition elements (MREs). Mechanistically, CCR492 functions as a competing endogenous RNA (ceRNA) to antagonize the function of let-7 microRNAs, leading to the de-repression of c-Myc. Moreover, we show that ectopic expression of CCR492 along with a constitutively active H-Ras promotes cell transformation. Thus, we identified a new lincRNA expressed as an early gene in mammalian cells to regulate the cell-cycle progression by upregulating c-Myc expression.


Subject(s)
Cell Transformation, Neoplastic/genetics , Fibroblasts/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Long Noncoding/genetics , Animals , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Embryo, Mammalian , Fibroblasts/cytology , G1 Phase , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Long Noncoding/metabolism , Transcriptional Activation
6.
Nucleic Acids Res ; 43(14): 6814-26, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-25925565

ABSTRACT

Ten-eleven translocation (Tet) genes encode for a family of hydroxymethylase enzymes involved in regulating DNA methylation dynamics. Tet1 is highly expressed in mouse embryonic stem cells (ESCs) where it plays a critical role the pluripotency maintenance. Tet1 is also involved in cell reprogramming events and in cancer progression. Although the functional role of Tet1 has been largely studied, its regulation is poorly understood. Here we show that Tet1 gene is regulated, both in mouse and human ESCs, by the stemness specific factors Oct3/4, Nanog and by Myc. Thus Tet1 is integrated in the pluripotency transcriptional network of ESCs. We found that Tet1 is switched off by cell proliferation in adult cells and tissues with a consequent genome-wide reduction of 5hmC, which is more evident in hypermethylated regions and promoters. Tet1 downmodulation is mediated by the Polycomb repressive complex 2 (PRC2) through H3K27me3 histone mark deposition. This study expands the knowledge about Tet1 involvement in stemness circuits in ESCs and provides evidence for a transcriptional relationship between Tet1 and PRC2 in adult proliferating cells improving our understanding of the crosstalk between the epigenetic events mediated by these factors.


Subject(s)
DNA-Binding Proteins/genetics , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Polycomb Repressive Complex 2/metabolism , Proto-Oncogene Proteins/genetics , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/metabolism , Down-Regulation , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mixed Function Oxygenases , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic
7.
Int J Cancer ; 139(2): 404-13, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26941084

ABSTRACT

Tumour progression depends on several sequential events that include the microenvironment remodelling processes and the switch to the angiogenic phenotype, leading to new blood vessels recruitment. Non-invasive imaging techniques allow the monitoring of functional alterations in tumour vascularity and cellularity. The aim of this work was to detect functional changes in vascularisation and cellularity through Dynamic Contrast Enhanced (DCE) and Diffusion Weighted (DW) Magnetic Resonance Imaging (MRI) modalities during breast cancer initiation and progression of a transgenic mouse model (BALB-neuT mice). Histological examination showed that BALB-neuT mammary glands undergo a slow neoplastic progression from simple hyperplasia to invasive carcinoma, still preserving normal parts of mammary glands. DCE-MRI results highlighted marked functional changes in terms of vessel permeability (K(trans) , volume transfer constant) and vascularisation (vp , vascular volume fraction) in BALB-neuT hyperplastic mammary glands if compared to BALB/c ones. When breast tissue progressed from simple to atypical hyperplasia, a strong increase in DCE-MRI biomarkers was observed in BALB-neuT in comparison to BALB/c mice (K(trans) = 5.3 ± 0.7E-4 and 3.1 ± 0.5E-4; vp = 7.4 ± 0.8E-2 and 4.7 ± 0.6E-2 for BALB-neuT and BALB/c, respectively) that remained constant during the successive steps of the neoplastic transformation. Consistent with DCE-MRI observations, microvessel counting revealed a significant increase in tumour vessels. Our study showed that DCE-MRI estimates can accurately detect the angiogenic switch at early step of breast cancer carcinogenesis. These results support the view that this imaging approach is an excellent tool to characterize microvasculature changes, despite only small portions of the mammary glands developed neoplastic lesions in a transgenic mouse model.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Contrast Media , Image Enhancement , Magnetic Resonance Imaging , Neovascularization, Pathologic/diagnostic imaging , Animals , Breast/pathology , Cell Transformation, Neoplastic , Disease Models, Animal , Female , Humans , Hyperplasia , Magnetic Resonance Imaging/methods , Mice , Mice, Transgenic
8.
Stem Cells ; 33(3): 742-50, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25504116

ABSTRACT

Although much is known about the pluripotency self-renewal circuitry, the molecular events that lead embryonic stem cells (ESCs) exit from pluripotency and begin differentiation are largely unknown. We found that the zinc finger transcription factor Snai1, involved in gastrulation and epithelial-mesenchymal transition, is already expressed in the inner cell mass of the preimplantation blastocysts. In ESCs, Snai1 does not respond to TGFß or BMP4 signaling but it is induced by retinoic acid treatment, which induces the binding, on the Snai1 promoter, of the retinoid receptors RARγ and RXRα, the dissociation of the Polycomb repressor complex 2 which results in the decrease of H3K27me3, and the increase of histone H3K4me3. Snai1 mediates the repression of pluripotency genes by binding directly to the promoters of Nanog, Nr5a2, Tcl1, c-Kit, and Tcfcp2l1. The transient activation of Snai1 in embryoid bodies induces the expression of the markers of all three germ layers. These results suggest that Snai1 is a key factor that triggers ESCs exit from the pluripotency state and initiate their differentiation processes.


Subject(s)
Embryonic Stem Cells/physiology , Pluripotent Stem Cells/physiology , Transcription Factors/genetics , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Nanog Homeobox Protein , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Tretinoin/pharmacology
9.
J Am Soc Nephrol ; 26(10): 2349-60, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25901032

ABSTRACT

Phenotypic changes induced by extracellular vesicles have been implicated in mesenchymal stromal cell-promoted recovery of AKI. MicroRNAs are potential candidates for cell reprogramming toward a proregenerative phenotype. The aim of this study was to evaluate whether microRNA deregulation inhibits the regenerative potential of mesenchymal stromal cells and derived extracellular vesicles in a model of glycerol-induced AKI in severe combined immunodeficient mice. We generated mesenchymal stromal cells depleted of Drosha to alter microRNA expression. Drosha-knockdown cells produced extracellular vesicles that did not differ from those of wild-type cells in quantity, surface molecule expression, and internalization within renal tubular epithelial cells. However, these vesicles showed global downregulation of microRNAs. Whereas wild-type mesenchymal stromal cells and derived vesicles administered intravenously induced morphologic and functional recovery in AKI, the Drosha-knockdown counterparts were ineffective. RNA sequencing analysis showed that kidney genes deregulated after injury were restored by treatment with mesenchymal stromal cells and derived vesicles but not with Drosha-knockdown cells and vesicles. Gene ontology analysis showed in AKI an association of downregulated genes with fatty acid metabolism and upregulated genes with inflammation, matrix-receptor interaction, and cell adhesion molecules. These alterations reverted after treatment with wild-type mesenchymal stromal cells and extracellular vesicles but not after treatment with the Drosha-knockdown counterparts. In conclusion, microRNA depletion in mesenchymal stromal cells and extracellular vesicles significantly reduced their intrinsic regenerative potential in AKI, suggesting a critical role of microRNAs in recovery after AKI.


Subject(s)
Acute Kidney Injury/therapy , Extracellular Vesicles , Mesenchymal Stem Cells/ultrastructure , MicroRNAs , Animals , Female , Mice
10.
J Exp Clin Cancer Res ; 42(1): 20, 2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36639824

ABSTRACT

BACKGROUND: Tumor progression is based on a close interaction between cancer cells and Tumor MicroEnvironment (TME). Here, we focus on the role that Cancer Associated Fibroblasts (CAFs), Mesenchymal Stem Cells (MSCs) and microRNAs (miRs) play in breast cancer and melanoma malignancy. METHODS: We used public databases to investigate miR-214 expression in the stroma compartment of primary human samples and evaluated tumor formation and dissemination following tumor cell injections in miR-214 overexpressing (miR-214over) and knock out (miR-214ko) mice. In addition, we dissected the impact of Conditioned Medium (CM) or Extracellular Vesicles (EVs) derived from miR-214-rich or depleted stroma cells on cell metastatic traits. RESULTS: We evidence that the expression of miR-214 in human cancer or metastasis samples mostly correlates with stroma components and, in particular, with CAFs and MSCs. We present data revealing that the injection of tumor cells in miR-214over mice leads to increased extravasation and metastasis formation. In line, treatment of cancer cells with CM or EVs derived from miR-214-enriched stroma cells potentiate cancer cell migration/invasion in vitro. Conversely, dissemination from tumors grown in miR-214ko mice is impaired and metastatic traits significantly decreased when CM or EVs from miR-214-depleted stroma cells are used to treat cells in culture. Instead, extravasation and metastasis formation are fully re-established when miR-214ko mice are pretreated with miR-214-rich EVs of stroma origin. Mechanistically, we also show that tumor cells are able to induce miR-214 production in stroma cells, following the activation of IL-6/STAT3 signaling, which is then released via EVs subsequently up-taken by cancer cells. Here, a miR-214-dependent pro-metastatic program becomes activated. CONCLUSIONS: Our findings highlight the relevance of stroma-derived miR-214 and its release in EVs for tumor dissemination, which paves the way for miR-214-based therapeutic interventions targeting not only tumor cells but also the TME.


Subject(s)
Breast Neoplasms , Mesenchymal Stem Cells , MicroRNAs , Humans , Animals , Mice , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction , Breast Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Stromal Cells/metabolism , Tumor Microenvironment
11.
FASEB J ; 24(8): 2680-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20354140

ABSTRACT

Loss of the fumarate hydratase (FH) tumor suppressor gene results in the development of benign tumors that rarely, but regrettably, progress to very aggressive cancers. Using mouse embryo fibroblasts (MEFs) to model transformation, we found that fh knockdown results in increased expression of the met oncogene-encoded tyrosine kinase receptor through hypoxia-inducible factor (hif) stabilization. MET-increased expression was alone able to stabilize hif, thus establishing a feed forward loop that might enforce tumor progression. The fh-defective MEFs showed increased motility and protection from apoptosis. Motility, but not survival, relied on hif-1alpha and was greatly enhanced by MET ligand hepatocyte growth factor. Met cooperated with a weakly oncogenic ras in making MEFs transformed and tumorigenic, as shown by in vitro and in vivo assays. Loss of fh was not equally effective by itself but enhanced the transformed and tumorigenic phenotype induced by ras and MET. Consistently, the rescue of fumarase expression abrogated the motogenic and transformed phenotype of fh-defective MEFs. In conclusion, the data suggest that the progression of tumors where FH is lost might be boosted by activation of the MET oncogene, which is able to drive cell-autonomous tumor progression and is a strong candidate for targeted therapy.


Subject(s)
Cell Transformation, Neoplastic , Fumarate Hydratase/physiology , Neoplasms/etiology , Proto-Oncogene Proteins c-met/physiology , Animals , Cells, Cultured , Fibroblasts , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Hepatocyte Growth Factor/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Tumor Suppressor Proteins/genetics
12.
Cancer Lett ; 510: 13-23, 2021 07 10.
Article in English | MEDLINE | ID: mdl-33862151

ABSTRACT

An interactive crosstalk between tumor and stroma cells is essential for metastatic melanoma progression. We evidenced that ESDN/DCBLD2/CLCP1 plays a crucial role in endothelial cells during the spread of melanoma. Precisely, increased extravasation and metastasis formation were revealed in ESDN-null mice injected with melanoma cells, even if the primary tumor growth, vessel permeability, and angiogenesis were not enhanced. Interestingly, improved adhesion of melanoma cells to ESDN-depleted endothelial cells was observed, due to the presence of higher levels of E-selectin transcripts/proteins in ESDN-defective cells. In accordance with these results, anticorrelation was observed between ESDN and E-selectin in human endothelial cells. Most importantly, our data revealed that cimetidine, an E-selectin inhibitor, was able to block cell adhesion, extravasation, and metastasis formation in ESDN-null mice, underlying a major role of ESDN in E-selectin transcription upregulation, which according to our data, may presumably be linked to STAT3. Based on our results, we propose a protective role for ESDN during the spread of melanoma and reveal its therapeutic potential.


Subject(s)
E-Selectin/antagonists & inhibitors , Endothelial Cells/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Disease Models, Animal , Disease Progression , E-Selectin/biosynthesis , E-Selectin/metabolism , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Tumor Microenvironment
13.
J Pathol ; 219(3): 356-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19718705

ABSTRACT

Vascular endothelial growth factor-D (VEGF-D) is one of the two ligands of the VEGFR-3 receptor on lymphatic endothelial cells. Gene-silencing studies in mice and Xenopus tadpoles recently showed that the role of endogenous VEGF-D in lymphatic development is moderate. By contrast, exogenous VEGF-D is capable of stimulating lymphangiogenesis. Nonetheless, its endogenous role in pathological conditions remains largely unknown. Hence, we reassessed its role in disease, using Vegf-d(null) mice. Vegf-d(null) mice were generated that, under physiological conditions, displayed normal embryonic and postnatal lymphangiogenesis and lymphatic remodelling, efficient lymphatic functioning and normal health. Vegf-d(null) mice also reponded normally in models of skin wound healing and healing of infarcted myocardium, despite enhanced expression of VEGF-D in these models in wild-type mice. In contrast, Vegf-d(null) mice displayed reduced peritumoral lymphangiogenesis and lymph node metastasis in an orthotopic pancreatic tumour model. Together, our data indicate that endogenous VEGF-D in mice is dispensible for lymphangiogenesis during development, in postnatal and adult physiology and in several pathological conditions, but significantly contributes to lymphatic metastasis.


Subject(s)
Lymphangiogenesis/physiology , Lymphatic Metastasis/physiopathology , Vascular Endothelial Growth Factor D/deficiency , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Animals , Disease Models, Animal , Embryonic Development/physiology , Female , Gene Targeting/methods , Liver Neoplasms/metabolism , Lymphangioma/metabolism , Lymphatic System/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/physiopathology , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Phenotype , Skin/injuries , Vascular Endothelial Growth Factor D/physiology , Wound Healing/physiology
14.
PLoS One ; 15(2): e0228062, 2020.
Article in English | MEDLINE | ID: mdl-32012171

ABSTRACT

Melatonin, a hormone secreted by pineal gland, exerts antimetastatic effects by reducing tumor cell proliferation, migration and invasion. MicroRNAs (miRNAs) are small, non-coding RNAs that play a crucial role in regulation of gene expression and biological processes of the cells. Herein, we search for a link between the tumor/metastatic-suppressive actions of melatonin and miRNA expression in triple-negative breast cancer cells. We demonstrated that melatonin exerts its anti-tumor actions by reducing proliferation, migration and c-Myc expression of triple negative breast cancer cells. By using Taqman-based assays, we analyzed the expression levels of a set of miRNAs following melatonin treatment of triple negative breast cancer cells and we identified 17 differentially expressed miRNAs, 6 down-regulated and 11 up-regulated. We focused on the anti-metastatic miR-148b and the oncogenic miR-210 both up-regulated by melatonin treatment and studied the effect of their modulation on melatonin-mediated impairment of tumor progression. Surprisingly, when miR-148b or miR-210 were depleted in triple-negative breast cancer cells, using a specific miR-148b sponge or anti-miR-210, melatonin effects on migration inhibition and c-myc downregulation were still visible suggesting that the increase of miR-148b and miR-210 expression observed following melatonin treatment was not required for the efficacy of melatonin action. Nevertheless, ours results suggest that melatonin exhibit a compound for metastatic trait inhibition, especially in MDA-MB-231 breast cancer cells even if a direct link between modulation of expression of certain proteins or miRNAs and melatonin effects has still to be established.


Subject(s)
Melatonin/pharmacology , MicroRNAs/genetics , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Proto-Oncogene Proteins c-myc/metabolism
15.
Clin Cancer Res ; 13(7): 2191-8, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17404103

ABSTRACT

PURPOSE: Advanced ovarian cancers are initially responsive to combinatorial chemotherapy with platinum drugs and taxanes but, in most cases, develop drug resistance. We recently showed that, in vitro, hepatocyte growth factor (HGF) enhances death of human ovarian cancer cell lines treated with cisplatin (CDDP) and paclitaxel. The present study addresses whether in vivo HGF makes ovarian carcinoma cells more responsive to these chemotherapeutics. EXPERIMENTAL DESIGN: Using Lentiviral vectors carrying the HGF transgene, we transduced SK-OV-3 and NIH:OVCAR-3 ovarian carcinoma cell lines to obtain stable autocrine and paracrine HGF receptor activation. In vitro, we assayed growth, motility, invasiveness, and the response to CDDP and paclitaxel of the HGF-secreting bulk unselected cell populations. In vivo, we tested the cytotoxic effects of the drugs versus s.c. tumors formed by the wild-type and HGF-secreting cells in immunocompromised mice. Tumor-bearing mice were treated with CDDP (i.p.) and paclitaxel (i.v.), combined in different schedules and doses. RESULTS: In vitro, HGF-secreting cells did not show altered proliferation rates and survival but were strongly sensitized to the death triggered by CDDP and paclitaxel, alone or in combination. In vivo, we found a therapeutic window in which autocrine/paracrine HGF made tumors sensitive to low doses of the drugs, which were ineffective on their own. CONCLUSIONS: These data provide the proof-of-concept that in vivo gene therapy with HGF might be competent in sensitizing ovarian cancer cells to conventional chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Genetic Therapy/methods , Hepatocyte Growth Factor/genetics , Ovarian Neoplasms/drug therapy , Animals , Apoptosis , Blotting, Western , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Flow Cytometry , Genetic Vectors , Hepatocyte Growth Factor/metabolism , Humans , In Vitro Techniques , Lentivirus/genetics , Mice , Ovarian Neoplasms/metabolism , Paclitaxel/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Transgenes
16.
Cancer Res ; 76(17): 5151-62, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27328731

ABSTRACT

miR-214 and miR-148b have been proposed to antagonize the effects of each other in enabling or blocking metastasis, respectively. In this study, we provide evidence deepening their role and interrelationship in the process of metastatic dissemination. Depleting miR-214 or elevating miR-148b blocked the dissemination of melanoma or breast cancer cells, an effect that could be accentuated by dual alteration. Mechanistic investigations indicated that dual alteration suppressed passage of malignant cells through the blood vessel endothelium by reducing expression of the cell adhesion molecules ITGA5 and ALCAM. Notably, transendothelial migration in vitro and extravasation in vivo impaired by singly alternating miR-214 or miR-148b could be overridden by overexpression of ITGA5 or ALCAM in the same tumor cells. In clinical specimens of primary breast cancer or metastatic melanoma, we found a positive correlation between miR-214 and ITGA5 or ALCAM along with an inverse correlation of miR-214 and miR-148b in the same specimens. Our findings define an antagonistic relationship of miR-214 and miR-148b in determining the dissemination of cancer cells via tumor-endothelial cell interactions, with possible implications for microRNA-mediated therapeutic interventions aimed at blocking cancer extravasation. Cancer Res; 76(17); 5151-62. ©2016 AACR.


Subject(s)
Breast Neoplasms/pathology , Melanoma/pathology , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Animals , Antigens, CD/biosynthesis , Breast Neoplasms/genetics , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Endothelial Cells/pathology , Female , Fetal Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Immunoblotting , Melanoma/genetics , Mice , Mice, Inbred NOD , Mice, SCID
17.
Oncotarget ; 5(15): 5992-6002, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25115388

ABSTRACT

CDT2/L2DTL/RAMP is one of the substrate receptors of the Cullin Ring Ubiquitin Ligase 4 that targets for ubiquitin mediated degradation a number of substrates, such as CDT1, p21 and CHK1, involved in the regulation of cell cycle and survival. Here we show that CDT2 depletion was alone able to induce the apoptotic death in 12/12 human cancer cell lines from different tissues, regardless of the mutation profile and CDT2 expression level. Cell death was associated to rereplication and to loss of CDT1 degradation. Conversely, CDT2 depletion did not affect non-transformed human cells, such as immortalized kidney, lung and breast cell lines, and primary cultures of endothelial cells and osteoblasts. The ectopic over-expression of an activated oncogene, such as the mutation-activated RAS or the amplified MET in non-transformed immortalized breast cell lines and primary human osteoblasts, respectively, made cells transformed in vitro, tumorigenic in vivo, and susceptible to CDT2 loss. The widespread effect of CDT2 depletion in different cancer cells suggests that CDT2 is not in a synthetic lethal interaction to a single specific pathway. CDT2 likely is a non-oncogene to which transformed cells become addicted because of their enhanced cellular stress, such as replicative stress and DNA damage.


Subject(s)
Neoplasms/enzymology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , DNA Replication , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Female , Human Umbilical Vein Endothelial Cells , Humans , Neoplasms/genetics , Neoplasms/pathology , Phenotype
18.
Mol Cell Biol ; 33(6): 1198-209, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23319049

ABSTRACT

To form three-dimensional capillary tubes, endothelial cells must establish contacts with the extracellular matrix that provides signals for their proliferation, migration, and differentiation. The transcription factor Fosl1 plays a key role in the vasculogenic and angiogenic processes as Fosl1 knockout embryos die with vascular defects in extraembryonic tissues. Here, we show that Fosl1(-/-) embryonic stem cells differentiate into endothelial cells but fail to correctly assemble into primitive capillaries and to form tube-like structures. FOSL1 silencing affects in vitro angiogenesis, increases cell adhesion, and decreases cell mobility of primary human endothelial cells (HUVEC). We further show that FOSL1 is a repressor of αv and ß3 integrin expression and that the down-modulation of αvß3 rescues the angiogenic phenotype in FOSL1-silenced HUVEC, while the ectopic expression of αvß3 alone reproduces the phenotypic alterations induced by FOSL1 knockdown. FOSL1 represses the transcription of both αv and ß3 integrin genes by binding together with JunD to their proximal promoter via the transcription factor SP1. These data suggest that FOSL1-dependent negative regulation of αvß3 expression on endothelial cells is required for endothelial assembly into vessel structures.


Subject(s)
Capillaries/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Integrin alphaVbeta3/physiology , Proto-Oncogene Proteins c-fos/physiology , Animals , Capillaries/cytology , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Line , Cell Movement/genetics , Down-Regulation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endothelium, Vascular/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Mice , Neovascularization, Pathologic/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Vitronectin/genetics , Vitronectin/metabolism
19.
Cancer Cell ; 22(2): 263-77, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22897855

ABSTRACT

The success of chemotherapy in cancer treatment is limited by scarce drug delivery to the tumor and severe side-toxicity. Prolyl hydroxylase domain protein 2 (PHD2) is an oxygen/redox-sensitive enzyme that induces cellular adaptations to stress conditions. Reduced activity of PHD2 in endothelial cells normalizes tumor vessels and enhances perfusion. Here, we show that tumor vessel normalization by genetic inactivation of Phd2 increases the delivery of chemotherapeutics to the tumor and, hence, their antitumor and antimetastatic effect, regardless of combined inhibition of Phd2 in cancer cells. In response to chemotherapy-induced oxidative stress, pharmacological inhibition or genetic inactivation of Phd2 enhances a hypoxia-inducible transcription factor (HIF)-mediated detoxification program in healthy organs, which prevents oxidative damage, organ failure, and tissue demise. Altogether, our study discloses alternative strategies for chemotherapy optimization.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Gene Targeting , Neoplasms/drug therapy , Procollagen-Proline Dioxygenase/metabolism , Alleles , Animals , Antineoplastic Agents/pharmacology , Antioxidants/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cisplatin/adverse effects , Cisplatin/therapeutic use , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Heart Diseases/chemically induced , Heart Diseases/pathology , Heart Diseases/prevention & control , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Mice , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity/drug effects , Procollagen-Proline Dioxygenase/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL