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











Publication year range
1.
Nat Cancer ; 5(8): 1206-1226, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38844817

ABSTRACT

Many individuals with cancer are resistant to immunotherapies. Here, we identify the gene encoding the pyrimidine salvage pathway enzyme cytidine deaminase (CDA) among the top upregulated metabolic genes in several immunotherapy-resistant tumors. We show that CDA in cancer cells contributes to the uridine diphosphate (UDP) pool. Extracellular UDP hijacks immunosuppressive tumor-associated macrophages (TAMs) through its receptor P2Y6. Pharmacologic or genetic inhibition of CDA in cancer cells (or P2Y6 in TAMs) disrupts TAM-mediated immunosuppression, promoting cytotoxic T cell entry and susceptibility to anti-programmed cell death protein 1 (anti-PD-1) treatment in resistant pancreatic ductal adenocarcinoma (PDAC) and melanoma models. Conversely, CDA overexpression in CDA-depleted PDACs or anti-PD-1-responsive colorectal tumors or systemic UDP administration (re)establishes resistance. In individuals with PDAC, high CDA levels in cancer cells correlate with increased TAMs, lower cytotoxic T cells and possibly anti-PD-1 resistance. In a pan-cancer single-cell atlas, CDAhigh cancer cells match with T cell cytotoxicity dysfunction and P2RY6high TAMs. Overall, we suggest CDA and P2Y6 as potential targets for cancer immunotherapy.


Subject(s)
Drug Resistance, Neoplasm , Immunotherapy , Uridine Diphosphate , Humans , Uridine Diphosphate/metabolism , Immunotherapy/methods , Drug Resistance, Neoplasm/immunology , Animals , Mice , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/drug therapy , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Cell Line, Tumor , Receptors, Purinergic P2/metabolism , Macrophages/immunology , Macrophages/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Tumor Microenvironment/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/drug therapy , Nucleotides/metabolism , Immune Tolerance , Programmed Cell Death 1 Receptor
3.
Oncotarget ; 8(49): 84671-84684, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29156675

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive form of brain cancer with poor prognosis. Cancer cells are characterized by a specific redox environment that adjusts metabolism to its specific needs and allows the tumor to grow and metastasize. As a consequence, cancer cells and especially GBM cells suffer from elevated oxidative pressure which requires antioxidant-defense and other sanitation enzymes to be upregulated. MTH1, which degrades oxidized nucleotides, is one of these defense enzymes and represents a promising cancer target. We found MTH1 expression levels elevated and correlated with GBM aggressiveness and discovered that siRNA knock-down or inhibition of MTH1 with small molecules efficiently reduced viability of patient-derived GBM cultures. The effect of MTH1 loss on GBM viability was likely mediated through incorporation of oxidized nucleotides and subsequent DNA damage. We revealed that MTH1 inhibition targets GBM independent of aggressiveness as well as potently kills putative GBM stem cells in vitro. We used an orthotopic zebrafish model to confirm our results in vivo and light-sheet microscopy to follow the effect of MTH1 inhibition in GBM in real time. In conclusion, MTH1 represents a promising target for GBM therapy and MTH1 inhibitors may also be effective in patients that suffer from recurring disease.

4.
Cancer Res ; 74(14): 3983-94, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24830720

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive clinical subtype accounting for up to 20% of all breast cancers, but its malignant determinants remain largely undefined. Here, we show that in TNBC the overexpression of Fra-1, a component of the transcription factor AP-1, offers prognostic potential. Fra-1 depletion or its heterodimeric partner c-Jun inhibits the proliferative and invasive phenotypes of TNBC cells in vitro. Similarly, RNAi-mediated attenuation of Fra-1 or c-Jun reduced cellular invasion in vivo in a zebrafish tumor xenograft model. Exploring the AP-1 cistrome and the AP-1-regulated transcriptome, we obtained insights into the transcriptional regulatory networks of AP-1 in TNBC cells. Among the direct targets identified for Fra-1/c-Jun involved in proliferation, adhesion, and cell-cell contact, we found that AP-1 repressed the expression of E-cadherin by transcriptional upregulation of ZEB2 to stimulate cell invasion. Overall, this work illuminates the pathways through which TNBC cells acquire invasive and proliferative properties.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Transcription Factor AP-1/metabolism , Transcription, Genetic , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Gene Knockdown Techniques , Homeodomain Proteins/metabolism , Humans , MAP Kinase Signaling System , Models, Biological , Neoplasm Invasiveness , Neoplasms, Basal Cell , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Repressor Proteins/metabolism , Transcription Factor AP-1/genetics , Transcriptome , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Zinc Finger E-box Binding Homeobox 2
5.
J Clin Invest ; 124(3): 1069-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24487586

ABSTRACT

Mutations of the tumor suppressor TP53 are present in many forms of human cancer and are associated with increased tumor cell invasion and metastasis. Several mechanisms have been identified for promoting dissemination of cancer cells with TP53 mutations, including increased targeting of integrins to the plasma membrane. Here, we demonstrate a role for the filopodia-inducing motor protein Myosin-X (Myo10) in mutant p53-driven cancer invasion. Analysis of gene expression profiles from 2 breast cancer data sets revealed that MYO10 was highly expressed in aggressive cancer subtypes. Myo10 was required for breast cancer cell invasion and dissemination in multiple cancer cell lines and murine models of cancer metastasis. Evaluation of a Myo10 mutant without the integrin-binding domain revealed that the ability of Myo10 to transport ß1 integrins to the filopodia tip is required for invasion. Introduction of mutant p53 promoted Myo10 expression in cancer cells and pancreatic ductal adenocarcinoma in mice, whereas suppression of endogenous mutant p53 attenuated Myo10 levels and cell invasion. In clinical breast carcinomas, Myo10 was predominantly expressed at the invasive edges and correlated with the presence of TP53 mutations and poor prognosis. These data indicate that Myo10 upregulation in mutant p53-driven cancers is necessary for invasion and that plasma-membrane protrusions, such as filopodia, may serve as specialized metastatic engines.


Subject(s)
Breast Neoplasms/metabolism , Lung Neoplasms/metabolism , Myosins/genetics , Tumor Suppressor Protein p53/genetics , Up-Regulation , Animals , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Shape , Female , Gene Expression , Humans , Integrins/metabolism , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Mice , Mice, Nude , Mutation, Missense , Myosins/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Transport , Pseudopodia/metabolism , Zebrafish
6.
Proc Natl Acad Sci U S A ; 111(7): 2560-5, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550283

ABSTRACT

The cellular response to hypoxia is regulated by hypoxia-inducible factor-1α and -2α (HIF-1α and -2α). We have discovered that filamin A (FLNA), a large cytoskeletal actin-binding protein, physically interacts with HIF-1α and promotes tumor growth and angiogenesis. Hypoxia induces a calpain-dependent cleavage of FLNA to generate a naturally occurring C-terminal fragment that accumulates in the cell nucleus. This fragment interacts with the N-terminal portion of HIF-1α spanning amino acid residues 1-390 but not with HIF-2α. In hypoxia this fragment facilitates the nuclear localization of HIF-1α, is recruited to HIF-1α target gene promoters, and enhances HIF-1α function, resulting in up-regulation of HIF-1α target gene expression in a hypoxia-dependent fashion. These results unravel an important mechanism that selectively regulates the nuclear accumulation and function of HIF-1α and potentiates angiogenesis and tumor progression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/physiology , Filamins/metabolism , Gene Expression Regulation, Neoplastic/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic/genetics , Animals , Chromatin Immunoprecipitation , Fluorescence , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Immunoprecipitation , Mice , Mice, SCID , RNA Interference , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism
7.
Nat Commun ; 4: 2129, 2013.
Article in English | MEDLINE | ID: mdl-23831851

ABSTRACT

Anti-platelet-derived growth factor (PDGF) drugs are routinely used in front-line therapy for the treatment of various cancers, but the molecular mechanism underlying their dose-dependent impact on vascular remodelling remains poorly understood. Here we show that anti-PDGF drugs significantly inhibit tumour growth and metastasis in high PDGF-BB-producing tumours by preventing pericyte loss and vascular permeability, whereas they promote tumour cell dissemination and metastasis in PDGF-BB-low-producing or PDGF-BB-negative tumours by ablating pericytes from tumour vessels. We show that this opposing effect is due to PDGF-ß signalling in pericytes. Persistent exposure of pericytes to PDGF-BB markedly downregulates PDGF-ß and inactivation of the PDGF-ß signalling decreases integrin α1ß1 levels, which impairs pericyte adhesion to extracellular matrix components in blood vessels. Our data suggest that tumour PDGF-BB levels may serve as a biomarker for selection of tumour-bearing hosts for anti-PDGF therapy and unsupervised use of anti-PDGF drugs could potentially promote tumour invasion and metastasis.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Fibrosarcoma/blood supply , Gene Expression Regulation, Neoplastic , Lung Neoplasms/blood supply , Neovascularization, Pathologic/genetics , Pericytes/pathology , Proto-Oncogene Proteins c-sis/genetics , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Becaplermin , Benzamides/pharmacology , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Cell Movement , Cell Proliferation/drug effects , Fibrosarcoma/drug therapy , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Gene Expression Profiling , Humans , Imatinib Mesylate , Integrin alpha1beta1/genetics , Integrin alpha1beta1/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mice , Neoplasm Metastasis , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Pericytes/drug effects , Pericytes/immunology , Piperazines/pharmacology , Proto-Oncogene Proteins c-sis/antagonists & inhibitors , Proto-Oncogene Proteins c-sis/immunology , Pyrimidines/pharmacology , Signal Transduction , Xenograft Model Antitumor Assays
8.
Birth Defects Res C Embryo Today ; 93(2): 182-93, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21671357

ABSTRACT

Most in vivo preclinical disease models are based on mouse and other mammalian systems. However, these rodent-based model systems have considerable limitations to recapitulate clinical situations in human patients. Zebrafish have been widely used to study embryonic development, behavior, tissue regeneration, and genetic defects. Additionally, zebrafish also provides an opportunity to screen chemical compounds that target a specific cell population for drug development. Owing to the availability of various genetically manipulated strains of zebrafish, immune privilege during early embryonic development, transparency of the embryos, and easy and precise setup of hypoxia equipment, we have developed several disease models in both embryonic and adult zebrafish, focusing on studying the role of angiogenesis in pathological settings. These zebrafish disease models are complementary to the existing mouse models, allowing us to study clinically relevant processes in cancer and nonmalignant diseases, which otherwise would be difficult to study in mice. For example, dissemination and invasion of single human or mouse tumor cells from the primary site in association with tumor angiogenesis can be studied under normoxia or hypoxia in zebrafish embryos. Hypoxia-induced retinopathy in the adult zebrafish recapitulates the clinical situation of retinopathy development in diabetic patients or age-related macular degeneration. These zebrafish disease models offer exciting opportunities to understand the mechanisms of disease development, progression, and development of more effective drugs for therapeutic intervention.


Subject(s)
Cardiovascular System/embryology , Diabetic Retinopathy/physiopathology , Disease Models, Animal , Lymphatic System/embryology , Macular Degeneration/physiopathology , Neoplasms/physiopathology , Neovascularization, Pathologic/physiopathology , Zebrafish , Animals , Animals, Genetically Modified , Cardiovascular System/anatomy & histology , Cell Hypoxia/physiology , Humans , Lymphatic System/anatomy & histology , Lymphatic System/physiology , Neovascularization, Pathologic/etiology , Regeneration/physiology , Signal Transduction/physiology , Species Specificity
9.
Nat Protoc ; 5(12): 1903-10, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21127484

ABSTRACT

Hypoxia-induced vascular responses, including angiogenesis, vascular remodeling and vascular leakage, significantly contribute to the onset, development and progression of retinopathy. However, until recently there were no appropriate animal disease models recapitulating adult retinopathy available. In this article, we describe protocols that create hypoxia-induced retinopathy in adult zebrafish. Adult fli1:EGFP zebrafish are placed in hypoxic water for 3-10 d and retinal neovascularization is analyzed using confocal microscopy. It usually takes 11 d to obtain conclusive results using the hypoxia-induced retinopathy model in adult zebrafish. This model provides a unique opportunity to study kinetically the development of retinopathy in adult animals using noninvasive protocols and to assess therapeutic efficacy of orally active antiangiogenic drugs.


Subject(s)
Diabetic Retinopathy/etiology , Disease Models, Animal , Hypoxia/complications , Neovascularization, Pathologic/pathology , Zebrafish , Animals , Green Fluorescent Proteins/metabolism , Microscopy, Fluorescence , Neovascularization, Pathologic/etiology
10.
Nat Protoc ; 5(12): 1911-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21127485

ABSTRACT

Hypoxia facilitates tumor invasion and metastasis by promoting neovascularization and co-option of tumor cells in the peritumoral vasculature, leading to dissemination of tumor cells into the circulation. However, until recently, animal models and imaging technology did not enable monitoring of the early events of tumor cell invasion and dissemination in living animals. We recently developed a zebrafish metastasis model to dissect the detailed events of hypoxia-induced tumor cell invasion and metastasis in association with angiogenesis at the single-cell level. In this model, fluorescent DiI-labeled human or mouse tumor cells are implanted into the perivitelline cavity of 48-h-old zebrafish embryos, which are subsequently placed in hypoxic water for 3 d. Tumor cell invasion, metastasis and pathological angiogenesis are detected under fluorescent microscopy in the living fish. The average experimental time for this model is 7 d. Our protocol offers a remarkable opportunity to study molecular mechanisms of hypoxia-induced cancer metastasis.


Subject(s)
Disease Models, Animal , Hypoxia/complications , Neoplasm Metastasis/pathology , Neovascularization, Pathologic/pathology , Zebrafish/embryology , Animals , Cell Line, Tumor , Embryo, Nonmammalian , Humans , Mice , Microscopy, Fluorescence , Neoplasm Invasiveness/physiopathology , Neovascularization, Pathologic/etiology
11.
Cell Cycle ; 9(5): 913-7, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20160500

ABSTRACT

Clinically detectable metastases represent an ultimate consequence of the metastatic cascade that consists of distinct processes including tumor cell invasion, dissemination, metastatic niche formation, and re-growth into a detectable metastatic mass. Although angiogenesis is known to promote tumor growth, its role in facilitating early events of the metastatic cascade remains poorly understood. We have recently developed a zebrafish tumor model that enables us to study involvement of pathological angiogenesis in tumor invasion, dissemination and metastasis. This non-invasive in vivo model allows detection of single malignant cell dissemination under both normoxia and hypoxia. Further, hypoxia-induced VEGF significantly facilitates tumor cell invasion and dissemination. These findings demonstrate that VEGF-induced pathological angiogenesis is essential for tumor dissemination and further corroborates potentially beneficial effects of clinically ongoing anti-VEGF drugs for the treatment of various malignancies.


Subject(s)
Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic , Animals , Hypoxia , Models, Animal , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
12.
Proc Natl Acad Sci U S A ; 106(46): 19485-90, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19887629

ABSTRACT

Mechanisms underlying pathological angiogenesis in relation to hypoxia in tumor invasion and metastasis remain elusive. Here, we have developed a zebrafish tumor model that allows us to study the role of pathological angiogenesis under normoxia and hypoxia in arbitrating early events of the metastatic cascade at the single cell level. Under normoxia, implantation of a murine T241 fibrosarcoma into the perivitelline cavity of developing embryos of transgenic fli1:EGFP zebrafish did not result in significant dissemination, invasion, and metastasis. In marked contrast, under hypoxia substantial tumor cells disseminated from primary sites, invaded into neighboring tissues, and metastasized to distal parts of the fish body. Similarly, expression of the hypoxia-regulated angiogenic factor, vascular endothelial growth factor (VEGF) to a high level resulted in tumor cell dissemination and metastasis, which correlated with increased tumor neovascularization. Inhibition of VEGF receptor signaling pathways by sunitinib or VEGFR2 morpholinos virtually completely ablated VEGF-induced tumor cell dissemination and metastasis. To the best of our knowledge, hypoxia- and VEGF-induced pathological angiogenesis in promoting tumor dissemination, invasion, and metastasis has not been described perviously at the single cell level. Our findings also shed light on molecular mechanisms of beneficial effects of clinically available anti-VEGF drugs for cancer therapy.


Subject(s)
Disease Models, Animal , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Neovascularization, Pathologic/pathology , Zebrafish , Animals , Animals, Genetically Modified , Cell Hypoxia , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , 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/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL