24 results on '"Carolina De La Torre"'
Search Results
2. Hepatic passaging of NRAS-mutant melanoma influences adhesive properties and metastatic pattern
- Author
-
Bianca Dietsch, Céline Weller, Carsten Sticht, Carolina de la Torre, Martin Kramer, Sergij Goerdt, Cyrill Géraud, and Sebastian A. Wohlfeil
- Subjects
Cancer Research ,Oncology ,Genetics - Abstract
Background Liver metastasis is a poor prognostic factor for treatment of advanced cutaneous melanoma with either immunotherapy or targeted therapies. In this study we focused on NRAS mutated melanoma, a cohort with high unmet clinical need. Methods WT31 melanoma was repeatedly passaged over the liver after intravenous injections five times generating the subline WT31_P5IV. The colonization of target organs, morphology, vascularization and the gene expression profiles of metastases were analyzed. Results After intravenous injection lung metastasis was significantly decreased and a trend towards increased liver metastasis was detected for WT31_P5IV as compared to parental WT31. Besides, the ratio of lung to liver metastases was significantly smaller. Histology of lung metastases revealed reduced proliferation of WT31_P5IV in relation to WT31 while both size and necrotic areas were unaltered. Liver metastases of both sublines showed no differences in vascularization, proliferation or necrosis. To identify tumor-intrinsic factors that altered the metastatic pattern of WT31_P5IV RNA sequencing was performed and revealed a differential regulation of pathways involved in cell adhesion. Ex vivo fluorescence imaging confirmed that initial tumor cell retention in the lungs was significantly reduced in WT31_P5IV in comparison to WT31. Conclusion This study demonstrates that tumor-intrinsic properties influencing the metastatic pattern of NRAS mutated melanoma are strongly affected by hepatic passaging and the hematogenous route tumor cells take. It has implications for the clinical setting as such effects might also occur during metastatic spread or disease progression in melanoma patients.
- Published
- 2023
- Full Text
- View/download PDF
3. Epigenetic inactivation of the 5-methylcytosine RNA methyltransferase NSUN7 is associated with clinical outcome and therapeutic vulnerability in liver cancer
- Author
-
Vanessa Ortiz-Barahona, Marta Soler, Veronica Davalos, Carlos A. García-Prieto, Maxime Janin, Fernando Setien, Irene Fernández-Rebollo, Joan J. Bech-Serra, Carolina De La Torre, Sonia Guil, Alberto Villanueva, Pei-Hong Zhang, Li Yang, Marco Guarnacci, Ulrike Schumann, Thomas Preiss, Ugne Balaseviciute, Robert Montal, Josep M. Llovet, and Manel Esteller
- Subjects
Cancer Research ,Oncology ,Molecular Medicine - Abstract
Background RNA modifications are important regulators of transcript activity and an increasingly emerging body of data suggests that the epitranscriptome and its associated enzymes are altered in human tumors. Methods Combining data mining and conventional experimental procedures, NSUN7 methylation and expression status was assessed in liver cancer cell lines and primary tumors. Loss-of-function and transfection-mediated recovery experiments coupled with RNA bisulfite sequencing and proteomics determined the activity of NSUN7 in downstream targets and drug sensitivity. Results In this study, the initial screening for genetic and epigenetic defects of 5-methylcytosine RNA methyltransferases in transformed cell lines, identified that the NOL1/NOP2/Sun domain family member 7 (NSUN7) undergoes promoter CpG island hypermethylation-associated with transcriptional silencing in a cancer-specific manner. NSUN7 epigenetic inactivation was common in liver malignant cells and we coupled bisulfite conversion of cellular RNA with next-generation sequencing (bsRNA-seq) to find the RNA targets of this poorly characterized putative RNA methyltransferase. Using knock-out and restoration-of-function models, we observed that the mRNA of the coiled-coil domain containing 9B (CCDC9B) gene required NSUN7-mediated methylation for transcript stability. Most importantly, proteomic analyses determined that CCDC9B loss impaired protein levels of its partner, the MYC-regulator Influenza Virus NS1A Binding Protein (IVNS1ABP), creating sensitivity to bromodomain inhibitors in liver cancer cells exhibiting NSUN7 epigenetic silencing. The DNA methylation-associated loss of NSUN7 was also observed in primary liver tumors where it was associated with poor overall survival. Interestingly, NSUN7 unmethylated status was enriched in the immune active subclass of liver tumors. Conclusion The 5-methylcytosine RNA methyltransferase NSUN7 undergoes epigenetic inactivation in liver cancer that prevents correct mRNA methylation. Furthermore, NSUN7 DNA methylation-associated silencing is associated with clinical outcome and distinct therapeutic vulnerability.
- Published
- 2023
- Full Text
- View/download PDF
4. Identification of liver‐derived bone morphogenetic protein (BMP)‐9 as a potential new candidate for treatment of colorectal cancer
- Author
-
H Gaitantzi, Nuh N. Rahbari, Andreas Teufel, Katja Breitkopf-Heinlein, C Cai, Johannes Betge, Norbert Gretz, Emrullah Birgin, Matthias P. Ebert, Carolina De La Torre, and Timo Itzel
- Subjects
Inhibitor of Differentiation Protein 1 ,animal structures ,Colorectal cancer ,In silico ,Bone Morphogenetic Protein 2 ,bone morphogenetic protein‐9 ,colorectal cancer ,Bone Morphogenetic Protein 4 ,Malignancy ,Bone morphogenetic protein ,Gene expression ,Growth Differentiation Factor 2 ,noggin ,medicine ,Organoid ,Humans ,Noggin ,business.industry ,ID1 ,Original Articles ,Cell Biology ,medicine.disease ,Intestinal epithelium ,Liver ,Bone Morphogenetic Proteins ,Colonic Neoplasms ,embryonic structures ,Cancer research ,Molecular Medicine ,Original Article ,Colorectal Neoplasms ,business ,Signal Transduction - Abstract
Colorectal cancer (CRC) is a high‐incidence malignancy worldwide which still needs better therapy options. Therefore, the aim of the present study was to investigate the responses of normal or malignant human intestinal epithelium to bone morphogenetic protein (BMP)‐9 and to find out whether the application of BMP‐9 to patients with CRC or the enhancement of its synthesis in the liver could be useful strategies for new therapy approaches. In silico analyses of CRC patient cohorts (TCGA database) revealed that high expression of the BMP‐target gene ID1, especially in combination with low expression of the BMP‐inhibitor noggin, is significantly associated with better patient survival. Organoid lines were generated from human biopsies of colon cancer (T‐Orgs) and corresponding non‐malignant areas (N‐Orgs) of three patients. The N‐Orgs represented tumours belonging to three different consensus molecular subtypes (CMS) of CRC. Overall, BMP‐9 stimulation of organoids promoted an enrichment of tumour‐suppressive gene expression signatures, whereas the stimulation with noggin had the opposite effects. Furthermore, treatment of organoids with BMP‐9 induced ID1 expression (independently of high noggin levels), while treatment with noggin reduced ID1. In summary, our data identify the ratio between ID1 and noggin as a new prognostic value for CRC patient outcome. We further show that by inducing ID1, BMP‐9 enhances this ratio, even in the presence of noggin. Thus, BMP‐9 is identified as a novel target for the development of improved anti‐cancer therapies of patients with CRC.
- Published
- 2021
- Full Text
- View/download PDF
5. YAP‐induced Ccl2 expression is associated with a switch in hepatic macrophage identity and vascular remodelling in liver cancer
- Author
-
Yingyue Tang, Fabian Rose, Carolina De La Torre, Teng Wei, Eduard Ryschich, Carsten Sticht, Peter Schirmacher, Stefan Thomann, Kai Breuhahn, Thomas Ritz, Hanno Glimm, Marcell Tóth, Claudia R. Ball, and S Weiler
- Subjects
CCR2 ,Carcinoma, Hepatocellular ,Kupffer Cells ,Receptors, CCR2 ,Angiogenesis ,Cell Cycle Proteins ,Vascular Remodeling ,Biology ,CCL2 ,Ligands ,Mice ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,CX3CR1 ,Tumor Microenvironment ,Animals ,Humans ,Chemokine CCL2 ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,PDGFB ,Hepatology ,Macrophages ,Liver Neoplasms ,YAP-Signaling Proteins ,HCCS ,030220 oncology & carcinogenesis ,Cancer research ,Transcription Factors - Abstract
BACKGROUND & AIM The development of hepatocellular carcinoma (HCC) is associated with the formation of communication networks leading to the recruitment of disease-modifying macrophages. However, how oncogenes in tumour cells control paracrine communication is not fully understood. METHODS Transgenic mice with liver-specific expression of the constitutively active yes-associated protein (YAPS127A ) or an orthotopic implantation model served as tumour models. FACS-sorted F4/80+ /CD11bdim /CD146- /retinoid- macrophages from healthy and tumour-bearing livers were used for transcriptomic profiling. Expression data of 242 human HCCs and a tissue microarray consisting of 91 HCCs and seven liver tissues were analyzed. RESULTS Screening of primary tumour cells expressing YAPS127A identified CC chemokine ligand 2 (Ccl2) as a macrophage chemoattractant, whose expression was regulated in a YAP/TEA domain family member 4 (TEAD4)-dependent manner. Ccl2 expression was associated with a loss of Kupffer cells (KCs) and an increase in immature macrophages (Mɸimm ) in hepatocarcinogenesis. Recruited Mɸimm were characterized by a lack of functional polarization (M0 signature) and high expression of the Ccl2 receptors C-C motif chemokine receptor 2 (Ccr2), C-X3-C motif chemokine receptor 1 (Cx3cr1) and pro-angiogenic platelet-derived growth factors (Pdgfa/Pdgfb). Mɸimm formed cellular clusters in the perivascular space, which correlated with vascular morphometric changes indicative for angiogenesis. In human HCCs, the M0 signature served as an identifier for poor clinical outcome and CCL2 correlated with YAP expression and vascular network formation. CONCLUSIONS In conclusion, YAP/TEAD4-regulated Ccl2 associates with perivascular recruitment of unpolarized Mɸimm and may contribute to a proangiogenic microenvironment in liver cancer.
- Published
- 2021
- Full Text
- View/download PDF
6. YAP Orchestrates Heterotypic Endothelial Cell Communication via HGF/c-MET Signaling in Liver Tumorigenesis
- Author
-
Carsten Sticht, Carolin Mogler, Olga Ermakova, Fabian Rose, Stefan Thomann, Eduard Ryschich, Carolina De La Torre, Daniel Kazdal, Kai Breuhahn, Claudia R. Ball, Norbert Gretz, Stephanie Roessler, Simone Marquard, Sarah Fritzsche, Peter Schirmacher, S Weiler, Teng Wei, Eugen Rempel, Hanno Glimm, and Marcell Tóth
- Subjects
0301 basic medicine ,Cancer Research ,Cell signaling ,Carcinoma, Hepatocellular ,C-Met ,Carcinogenesis ,Cell ,Paracrine Communication ,Cell Cycle Proteins ,Mice, Transgenic ,Cell Communication ,Biology ,medicine.disease_cause ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Humans ,Adaptor Proteins, Signal Transducing ,Oncogene ,Hepatocyte Growth Factor ,Liver Neoplasms ,Endothelial Cells ,YAP-Signaling Proteins ,Hep G2 Cells ,Proto-Oncogene Proteins c-met ,Mice, Inbred C57BL ,Endothelial stem cell ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Hepatocyte growth factor ,Signal Transduction ,Transcription Factors ,medicine.drug - Abstract
The oncogene yes-associated protein (YAP) controls liver tumor initiation and progression via cell extrinsic functions by creating a tumor-supporting environment in conjunction with cell autonomous mechanisms. However, how YAP controls organization of the microenvironment and in particular the vascular niche, which contributes to liver disease and hepatocarcinogenesis, is poorly understood. To investigate heterotypic cell communication, we dissected murine and human liver endothelial cell (EC) populations into liver sinusoidal endothelial cells (LSEC) and continuous endothelial cells (CEC) through histomorphological and molecular characterization. In YAPS127A-induced tumorigenesis, a gradual replacement of LSECs by CECs was associated with dynamic changes in the expression of genes involved in paracrine communication. The formation of new communication hubs connecting CECs and LSECs included the hepatocyte growth factor (Hgf)/c-Met signaling pathway. In hepatocytes and tumor cells, YAP/TEA domain transcription factor 4 (TEAD4)–dependent transcriptional induction of osteopontin (Opn) stimulated c-Met expression in EC with CEC phenotype, which sensitized these cells to the promigratory effects of LSEC-derived Hgf. In human hepatocellular carcinoma, the presence of a migration-associated tip-cell signature correlated with poor clinical outcome and the loss of LSEC marker gene expression. The occurrence of c-MET–expressing CECs in human liver cancer samples was confirmed at the single-cell level. In summary, YAP-dependent changes of the liver vascular niche comprise the formation of heterologous communication hubs in which tumor cell–derived factors modify the cross-talk between LSECs and CECs via the HGF/c-MET axis. Significance: YAP-dependent changes of the liver vascular niche comprise the formation of heterologous communication hubs in which tumor cell-derived factors modify the cross-talk between EC subpopulations.
- Published
- 2020
- Full Text
- View/download PDF
7. Yes-associated protein (YAP) induces a secretome phenotype and transcriptionally regulates plasminogen activator Inhibitor-1 (PAI-1) expression in hepatocarcinogenesis
- Author
-
Peter Schirmacher, Stefan Thomann, S Weiler, Carsten Sticht, Beate K. Straub, Jens U. Marquardt, Simone Marquard, Norbert Gretz, Marcell Tóth, Kai Breuhahn, Carolina De La Torre, Teresa Lutz, and Michaela Bissinger
- Subjects
Proteome ,Transcription, Genetic ,Carcinogenesis ,Hepatocellular carcinoma ,Hippo pathway ,Muscle Proteins ,lcsh:Medicine ,Tumor initiation ,Biochemistry ,0302 clinical medicine ,Promoter Regions, Genetic ,Cellular Senescence ,0303 health sciences ,lcsh:Cytology ,Liver Neoplasms ,TEA Domain Transcription Factors ,Prognosis ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Phenotype ,Transcriptional regulator ,030220 oncology & carcinogenesis ,CYR61 ,Liver cancer ,Carcinoma, Hepatocellular ,Mice, Transgenic ,Biology ,03 medical and health sciences ,Cell Line, Tumor ,Plasminogen Activator Inhibitor 1 ,Gene silencing ,Animals ,lcsh:QH573-671 ,Molecular Biology ,Transcription factor ,CXCL16 ,Oncogene ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Tumor microenvironment ,Research ,lcsh:R ,YAP-Signaling Proteins ,Cell Biology ,Disease Models, Animal ,Tumor progression ,Cancer research ,Hepatocytes ,Transcription Factors - Abstract
Background Overexpression and nuclear enrichment of the oncogene yes-associated protein (YAP) cause tumor initiation and support tumor progression in human hepatocellular carcinoma (HCC) via cell autonomous mechanisms. However, how YAP expression in tumor cells affects intercellular communication within the tumor microenvironment is not well understood. Methods To investigate how tumor cell-derived YAP is changing the paracrine communication network between tumor cells and non-neoplastic cells in hepatocarcinogenesis, the expression and secretion of cytokines, growth factors and chemokines were analyzed in transgenic mice with liver-specific and inducible expression of constitutively active YAP (YAPS127A). Transcriptomic and proteomic analyses were performed using primary isolated hepatocytes and blood plasma. In vitro, RNAinterference (RNAi), expression profiling, functional analyses and chromatin immunoprecipitation (ChIP) analyses of YAP and the transcription factor TEA domain transcription factor 4 (TEAD4) were performed using immortalized cell lines. Findings were confirmed in cohorts of HCC patients at the transcript and protein levels. Results YAP overexpression induced the expression and secretion of many paracrine-acting factors with potential impact on tumorous or non-neoplastic cells (e.g. plasminogen activator inhibitor-1 (PAI-1), C-X-C motif chemokine ligand 13 (CXCL13), CXCL16). Expression analyses of human HCC patients showed an overexpression of PAI-1 in human HCC tissues and a correlation with poor overall survival as well as early cancer recurrence. PAI-1 statistically correlated with genes typically induced by YAP, such as connective tissue growth factor (CTGF) and cysteine rich angiogenic inducer 61 (CYR61) or YAP-dependent gene signatures (CIN4/25). In vitro, YAP inhibition diminished the expression and secretion of PAI-1 in murine and human liver cancer cell lines. PAI-1 affected the expression of genes involved in cellular senescence and oncogene-induced senescence was confirmed in YAPS127A transgenic mice. Silencing of TEAD4 as well as treatment with the YAP/TEAD interfering substance Verteporfin reduced PAI-1 expression. ChIP analyses confirmed the binding of YAP and TEAD4 to the gene promoter of PAI-1 (SERPINE1). Conclusions These results demonstrate that the oncogene YAP changes the secretome response of hepatocytes and hepatocyte-derived tumor cells. In this context, the secreted protein PAI-1 is transcriptionally regulated by YAP in hepatocarcinogenesis. Perturbation of these YAP-dependent communication hubs including PAI-1 may represent a promising pharmacological approach in tumors with YAP overexpression.
- Published
- 2020
- Full Text
- View/download PDF
8. STAT3 inhibitor Napabucasin abrogates MDSC immunosuppressive capacity and prolongs survival of melanoma-bearing mice
- Author
-
Rebekka Bitsch, Annina Kurzay, Feyza Özbay Kurt, Carolina De La Torre, Samantha Lasser, Alisa Lepper, Alina Siebenmorgen, Verena Müller, Peter Altevogt, Jochen Utikal, and Viktor Umansky
- Subjects
Pharmacology ,STAT3 Transcription Factor ,Cancer Research ,Myeloid-Derived Suppressor Cells ,Immunology ,Mice, Transgenic ,Mice ,Oncology ,Molecular Medicine ,Immunology and Allergy ,Animals ,Humans ,Melanoma ,Benzofurans ,Naphthoquinones - Abstract
BackgroundMyeloid-derived suppressor cells (MDSCs) represent a negative prognostic factor in malignant melanoma. These cells are generated under chronic inflammatory conditions typical of cancer. The transcription factor signal transducer and activator of transcription 3 (STAT3) orchestrates MDSC accumulation and acquisition of immunosuppressive properties. Here we studied STAT3 inhibition by Napabucasin as a way to block MDSC accumulation and activity and its potential to treat malignant melanoma.MethodsIn vitro generated murine MDSC and primary MDSC from melanoma-bearing mice were used to investigate the effects of Napabucasin on MDSC in vitro. The RET transgenic mouse model of malignant melanoma was used to examine Napabucasin therapy efficiency and its underlying mechanisms in vivo. Furthermore, STAT3 activation and its correlation with survival were explored in MDSC from 19 patients with malignant melanoma and human in vitro generated monocytic myeloid-derived suppressor cell (M-MDSC) were used to evaluate the effects of Napabucasin.ResultsNapabucasin was able to abrogate the capacity of murine MDSC to suppress CD8+ T-cell proliferation. The STAT3 inhibitor induced apoptosis in murine MDSC, significantly increased expression of molecules associated with antigen processing and presentation, as well as slightly decreased expression of immunosuppressive factors on these cells. RET transgenic mice treated with Napabucasin showed prolonged survival accompanied by a strong accumulation of tumor-infiltrating antigen-presenting cells and activation of CD8+ and CD4+ T cells. Interestingly, patients with malignant melanoma with high expression of activated STAT3 in circulating M-MDSC showed significantly worse progression-free survival (PFS) than patients with low levels of activated STAT3. In addition, Napabucasin was able to abrogate suppressive capacity of human in vitro generated M-MDSC.ConclusionOur findings demonstrate that STAT3 inhibitor Napabucasin completely abrogated the immunosuppressive capacity of murine MDSC and human M-MDSC and improved melanoma-bearing mouse survival. Moreover, patients with malignant melanoma with high expression levels of activated STAT3 in M-MDSC displayed shorter PFS, indicating its role as a promising therapeutic target in patients with malignant melanoma and a predictive marker for their clinical outcome.
- Published
- 2022
9. Establishment of Tumor Treating Fields Combined With Mild Hyperthermia as Novel Supporting Therapy for Pancreatic Cancer
- Author
-
Shuyang Zhao, Wolfgang Gross, Tobias Pfeifer, Ingrid Herr, Carolina De La Torre, Li Liu, Liping Bai, and Michael Schaefer
- Subjects
Hyperthermia ,Pancreatic duct ,Cancer Research ,Microarray ,business.industry ,DNA repair ,Autophagy ,pancreatic ductal adenocarcinoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell cycle ,hyperthermia ,bioinformatics and computational biology ,medicine.disease ,alternative therapies ,tumor treating fields ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,KEGG ,business ,RC254-282 ,Original Research - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with poor prognosis and limited therapeutic options. Alternating electrical fields with low intensity called “Tumor Treating Fields” (TTFields) are a new, non-invasive approach with almost no side effects and phase 3 trials are ongoing in advanced PDAC. We evaluated TTFields in combination with mild hyperthermia. Three established human PDAC cell lines and an immortalized pancreatic duct cell line were treated with TTFields and hyperthermia at 38.5°C, followed by microscopy, assays for MTT, migration, colony and sphere formation, RT-qPCR, FACS, Western blot, microarray and bioinformatics, and in silico analysis using the online databases GSEA, KEGG, Cytoscape-String, and Kaplan-Meier Plotter. Whereas TTFields and hyperthermia alone had weak effects, their combination strongly inhibited the viability of malignant, but not those of nonmalignant cells. Progression features and the cell cycle were impaired, and autophagy was induced. The identified target genes were key players in autophagy, the cell cycle and DNA repair. The expression profiles of part of these target genes were significantly involved in the survival of PDAC patients. In conclusion, the combination of TTFields with mild hyperthermia results in greater efficacy without increased toxicity and could be easily clinically approved as supporting therapy.
- Published
- 2021
- Full Text
- View/download PDF
10. N-Octanoyl-Dopamine inhibits cytokine production in activated T-cells and diminishes MHC-class-II expression as well as adhesion molecules in IFNγ-stimulated endothelial cells
- Author
-
Benito A. Yard, Carolina De La Torre, Nicolas Krapp, Anna-Isabelle Kälsch, Bernhard K. Krämer, Marloes Sol, M Kolibabka, Prama Pallavi, Björn B. Hofmann, Jana D. Braun, and Yingchun Li
- Subjects
0301 basic medicine ,CD74 ,Dopamine ,T-Lymphocytes ,medicine.medical_treatment ,lcsh:Medicine ,Nod ,Integrin alpha4beta1 ,030230 surgery ,Kidney ,Lymphocyte Activation ,0302 clinical medicine ,lcsh:Science ,Kidney diseases ,Multidisciplinary ,IMMUNOSUPPRESSION ,biology ,BRAIN-DEATH ,Cell adhesion molecule ,Chemistry ,PROLIFERATION ,Nuclear Proteins ,Intercellular Adhesion Molecule-1 ,HLA-DR MOLECULES ,Lymphocyte Function-Associated Antigen-1 ,Cytokine ,REJECTION ,Tumor necrosis factor alpha ,Signal Transduction ,KIDNEY-TRANSPLANTATION ,Vascular Cell Adhesion Molecule-1 ,CD18 ,chemical and pharmacologic phenomena ,Article ,Interferon-gamma ,03 medical and health sciences ,INFLAMMATION ,Cell Adhesion ,Human Umbilical Vein Endothelial Cells ,medicine ,CIITA ,Humans ,PRESERVATION ,ELIMINATION ,MHC class II ,lcsh:R ,Histocompatibility Antigens Class II ,HLA-DR Antigens ,Antigens, Differentiation, B-Lymphocyte ,030104 developmental biology ,Gene Expression Regulation ,Trans-Activators ,biology.protein ,Cancer research ,lcsh:Q ,Cell Adhesion Molecules ,GENERATION - Abstract
IFNγ enhances allograft immunogenicity and facilitates T-cell mediated rejection. This may cause interstitial fibrosis and tubular atrophy (IFTA), contributing to chronic allograft loss. We assessed if inhibition of T-cell activation by N-octanoyl dopamine (NOD) impairs adherence of activated T-cells to endothelial cells and the ability of activated T-cells to produce IFNγ. We also assessed if NOD affects IFNγ mediated gene expression in endothelial cells. The presence of NOD during T-cell activation significantly blunted their adhesion to unstimulated and cytokine stimulated HUVEC. Supernatants of these T-cells displayed significantly lower concentrations of TNFα and IFNγ and were less capable to facilitate T-cell adhesion. In the presence of NOD VLA-4 (CD49d/CD29) and LFA-1 (CD11a/CD18) expression on T-cells was reduced. NOD treatment of IFNγ stimulated HUVEC reduced the expression of MHC class II transactivator (CIITA), of MHC class II and its associated invariant chain CD74. Since IFTA is associated with T-cell mediated rejection and IFNγ to a large extent regulates immunogenicity of allografts, our current data suggest a potential clinical use of NOD in the treatment of transplant recipients. Further in vivo studies are warranted to confirm these in vitro findings and to assess the benefit of NOD on IFTA in clinically relevant models.
- Published
- 2019
- Full Text
- View/download PDF
11. ADP secreted by dying melanoma cells mediates chemotaxis and chemokine secretion of macrophages via the purinergic receptor P2Y12
- Author
-
Jochen Utikal, Carsten Sticht, Loreen Kloss, Kai Schledzewski, Viktor Umansky, Andreas Krewer, Calin Manta, Claudia Dollt, Astrid Schmieder, Susanne Melchers, and Carolina De La Torre
- Subjects
Ticagrelor ,Cancer Research ,Chemokine ,MAP Kinase Signaling System ,Chemokine CXCL2 ,Immunology ,Article ,Dexamethasone ,Monocytes ,Cellular and Molecular Neuroscience ,P2Y12 ,Cell Line, Tumor ,Humans ,Interleukin 8 ,Phosphorylation ,lcsh:QH573-671 ,Melanoma ,Phagocytes ,U937 cell ,biology ,Chemistry ,lcsh:Cytology ,Chemotaxis ,Macrophage Colony-Stimulating Factor ,Macrophages ,Interleukin-8 ,Purinergic receptor ,Cell Biology ,beta-Thromboglobulin ,Adenosine Monophosphate ,Receptors, Purinergic P2Y12 ,Clopidogrel ,Adenosine Diphosphate ,Gene Expression Regulation, Neoplastic ,CXCL2 ,Chemokine secretion ,biology.protein ,Cancer research ,Interleukin-4 ,Chemokines ,Prasugrel Hydrochloride - Abstract
Melanoma immunotherapy is still not satisfactory due to immunosuppressive cell populations within the tumor stroma. Targeting tumor-associated macrophages (TAM) can help to restore an anti-tumor immunity. Previously, we could show that classical TAM markers expressed in vivo need a 7 day M-CSF/dexamethasone/IL-4 (MDI) stimulation for their induction in peripheral blood monocytes (pBM) in vitro. To identify possible novel therapeutic targets on TAM, gene expression analysis of MDI-treated pBM was performed. This identified up-regulation of the purinergic G-protein coupled receptor P2Y12, the therapeutic target of the clinically approved anti-thrombotic drugs cangrelor, clopidogrel, ticagrelor, and prasugrel. We generated a peptide antibody and validated its specificity using transgenic P2Y12+ U937 cells. With the help of this antibody, P2Y12 expression was confirmed on CD68+ CD163+ TAM of melanoma in situ. Functional analysis revealed that treatment of transgenic P2Y12+ U937 cells with the receptor agonist 2-MeSADP induced ERK1/2 and Akt phosphorylation and increased the secretion of the chemokines CXCL2, CXCL7, and CXCL8. These effects could be abolished with the P2Y12 antagonist PSB0739 or with Akt and ERK inhibitors. In addition, P2Y12+ macrophages migrated towards the ADP-rich culture medium of puromycin-treated dying B16F1 melanoma cells. Cangrelor treatment blocked migration. Taken together, our results indicate that P2Y12 is an important chemotaxis receptor, which triggers migration of macrophages towards nucleotide-rich, necrotic tumor areas, and modulates the inflammatory environment upon ADP binding.
- Published
- 2019
- Full Text
- View/download PDF
12. YAP-dependent induction of UHMK1 supports nuclear enrichment of the oncogene MYBL2 and proliferation in liver cancer cells
- Author
-
Stephan Singer, Teng Wei, Jens U. Marquardt, Carsten Sticht, Stefan Thomann, Kai Breuhahn, Peter Schirmacher, Carolina De La Torre, Norbert Gretz, Thomas Ruppert, Teresa Lutz, Sabine Merker, S Weiler, Marcell Tóth, and Beate K. Straub
- Subjects
DNA Replication ,0301 basic medicine ,Cancer Research ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Biology ,Proto-Oncogene Mas ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,Humans ,MYB ,E2F ,Molecular Biology ,Mitosis ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Cell Nucleus ,Oncogene ,Cell growth ,Cell Cycle ,Liver Neoplasms ,Intracellular Signaling Peptides and Proteins ,YAP-Signaling Proteins ,Cell cycle ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Trans-Activators ,FOXM1 ,Protein Binding ,Transcription Factors - Abstract
The oncogene yes-associated protein (YAP) is a key modifier of liver homeostasis and regulates mitosis in hepatocytes as well as in malignantly transformed cells. However, the question of how YAP supports cell proliferation in hepatocellular carcinoma (HCC) is not well understood. Here we identified U2AF momology motif kinase 1 (UHMK1) as a direct transcriptional target of YAP and the transcription factor forkhead box M1 (FOXM1), which supports HCC cell proliferation but not migration. Indeed, UHMK1 stimulates the expression of genes that are specific for cell cycle regulation and which are known downstream effectors of YAP. By using BioID labeling and mass spectrometry, the dimerization partner, RB-like, E2F and multi-vulval class B (DREAM) complex constituent MYB proto-oncogene like 2 (MYBL2, B-MYB) was identified as a direct UHMK1 interaction partner. Like YAP, UHMK1 stimulates nuclear enrichment of MYBL2, which is associated HCC cell proliferation and the expression of the cell cycle regulators CCNB1, CCNB2, KIF20A, and MAD2L1. The association between YAP, UHMK1, MYBL2, and proliferation was confirmed in YAPS127A-transgenic mice and human HCC tissues. In summary, we provide a model by which YAP supports cell proliferation through the induction of important cell cycle regulators in a UHMK1- and MYBL2-dependent manner.
- Published
- 2019
- Full Text
- View/download PDF
13. Antitumor Activity of the Novel BTK Inhibitor TG-1701 Is Associated with Disruption of Ikaros Signaling in Patients with B-cell Non-Hodgkin Lymphoma
- Author
-
Núria Profitós Pelejà, Carolina De La Torre, Marcelo Lima Ribeiro, Marc Armengol, Pablo Menendez, Diana Reyes-Garau, Eva Musulen, Emmanuel Normant, Francesc Bosch, Juliana Carvalho Santos, H.P. Miskin, Meritxell Vinyoles, Miranda Fernández-Serrano, Alícia Sedó Mor, Gaël Roué, Pedro Blecua, Joan Josep Bech-Serra, and Manel Esteller
- Subjects
Cancer Research ,YES1 ,Chronic lymphocytic leukemia ,Mice ,Piperidines ,In vivo ,hemic and lymphatic diseases ,medicine ,Agammaglobulinaemia Tyrosine Kinase ,Bruton's tyrosine kinase ,Animals ,Humans ,Protein Kinase Inhibitors ,biology ,business.industry ,Lymphoma, Non-Hodgkin ,Gene signature ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Lymphoma ,Pyrimidines ,Oncology ,Cancer research ,biology.protein ,Biomarker (medicine) ,Pyrazoles ,Signal transduction ,business ,Signal Transduction - Abstract
Purpose: Despite the remarkable activity of BTK inhibitors (BTKi) in relapsed B-cell non–Hodgkin lymphoma (B-NHL), no clinically-relevant biomarker has been associated to these agents so far. The relevance of phosphoproteomic profiling for the early identification of BTKi responders remains underexplored. Experimental Design: A set of six clinical samples from an ongoing phase I trial dosing patients with chronic lymphocytic leukemia (CLL) with TG-1701, a novel irreversible and highly specific BTKi, were characterized by phosphoproteomic and RNA sequencing (RNA-seq) analysis. The activity of TG-1701 was evaluated in a panel of 11 B-NHL cell lines and mouse xenografts, including two NF-κB- and BTKC481S-driven BTKi-resistant models. Biomarker validation and signal transduction analysis were conducted through real-time PCR, Western blot analysis, immunostaining, and gene knockout (KO) experiments. Results: A nonsupervised, phosphoproteomic-based clustering did match the early clinical outcomes of patients with CLL and separated a group of “early-responders” from a group of “late-responders.” This clustering was based on a selected list of 96 phosphosites with Ikaros-pSer442/445 as a potential biomarker for TG-1701 efficacy. TG-1701 treatment was further shown to blunt Ikaros gene signature, including YES1 and MYC, in early-responder patients as well as in BTKi-sensitive B-NHL cell lines and xenografts. In contrast, Ikaros nuclear activity and signaling remained unaffected by the drug in vitro and in vivo in late-responder patients and in BTKC481S, BTKKO, and noncanonical NF-κB models. Conclusions: These data validate phosphoproteomic as a valuable tool for the early detection of response to BTK inhibition in the clinic, and for the determination of drug mechanism of action.
- Published
- 2021
14. Sulforaphane Inhibits the Expression of Long Noncoding RNA H19 and its Target APOBEC3G and Thereby Pancreatic Cancer Progression
- Author
-
Li Liu, Libo Yin, Huihui Ji, Zhimin Qi, Carolina De La Torre, Jury Gladkich, Ingrid Herr, Xuefeng An, Yiqiao Luo, and Bin Yan
- Subjects
0301 basic medicine ,TGF-β ,Cancer Research ,Small interfering RNA ,Xenotransplantation ,medicine.medical_treatment ,lncRNA-H19 ,pancreatic ductal adenocarcinoma ,sulforaphane ,In situ hybridization ,Biology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Pancreatic cancer ,medicine ,long noncoding RNAs ,APOBEC3G ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Long non-coding RNA ,Blot ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Sulforaphane - Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with poor therapeutic responses and short survival. The identification of factors that make PDAC so deadly and their targeting is important. Sulforaphane has shown promise in experimental and epidemiological studies, as well as in initial patient pilot studies. We examined the influence of sulforaphane to the largely unexplored epigenetic regulators “long noncoding RNAs” (lncRNAs). Sulforaphane modulated the expression of the lncRNAs H19, MALAT1, HOTAIR, HOTTIP and PVT1. The downregulation of the tumor promoter H19 and its target gene APOBEC3G was most significant and converged in inhibition of Smad2/TGF-β, which is involved in prevention of PDAC progression. Our data identified APOBEC3G as a new H19 target and a novel therapeutic target in PDAC, which can be inhibited by sulforaphane. The provided gene array accession numbers are important for future exploration of the suggested mechanism. Abstract Pancreatic ductal adenocarcinoma (PDAC) is extremely malignant and the therapeutic options available usually have little impact on survival. Great hope is placed on new therapeutic targets, including long noncoding RNAs (lncRNAs), and on the development of new drugs, based on e.g., broccoli-derived sulforaphane, which meanwhile has shown promise in pilot studies in patients. We examined whether sulforaphane interferes with lncRNA signaling and analyzed five PDAC and two nonmalignant cell lines, patient tissues (n = 30), and online patient data (n = 350). RT-qPCR, Western blotting, MTT, colony formation, transwell and wound healing assays; gene array analysis; bioinformatics; in situ hybridization; immunohistochemistry and xenotransplantation were used. Sulforaphane regulated the expression of all of five examined lncRNAs, but basal expression, biological function and inhibition of H19 were of highest significance. H19 siRNA prevented colony formation, migration, invasion and Smad2 phosphorylation. We identified 103 common sulforaphane- and H19-related target genes and focused to the virus-induced tumor promoter APOBEC3G. APOBEC3G siRNA mimicked the previously observed H19 and sulforaphane effects. In vivo, sulforaphane- or H19 or APOBEC3G siRNAs led to significantly smaller tumor xenografts with reduced expression of Ki67, APOBEC3G and phospho-Smad2. Together, we identified APOBEC3G as H19 target, and both are inhibited by sulforaphane in prevention of PDAC progression.
- Published
- 2021
15. Inhibition of CBP synergizes with the RNA-dependent mechanisms of Azacitidine by limiting protein synthesis
- Author
-
Antonio Gentilella, Johannes Zuber, Marguerite-Marie Le Pannérer, Carolina De La Torre, Michael Maher, René Winkler, Carolina Martínez Herráez, Lurdes Zamora, Joan Josep Bech-Serra, Matthias Muhar, Nigel Brooks, Marcus Buschbeck, Katharina Götze, Michaela Fellner, Mark van der Garde, Philipp Rathert, Raquel Casquero, and Jeannine Diesch
- Subjects
Antimetabolites, Antineoplastic ,Science ,Azacitidine ,Regulator ,General Physics and Astronomy ,Decitabine ,Therapeutics ,Leukemia, Myelomonocytic, Acute ,Article ,Acute myeloid leukaemia ,General Biochemistry, Genetics and Molecular Biology ,Small hairpin RNA ,Cell Line, Tumor ,Hematopoesi ,medicine ,Protein biosynthesis ,Humans ,heterocyclic compounds ,CREB-binding protein ,neoplasms ,Multidisciplinary ,biology ,Chemistry ,RNA ,General Chemistry ,DNA Methylation ,Terapèutica ,CREB-Binding Protein ,Hematopoiesis ,stomatognathic diseases ,Histone ,Protein Biosynthesis ,biology.protein ,Cancer research ,Myelodysplastic syndrome ,medicine.drug - Abstract
The nucleotide analogue azacitidine (AZA) is currently the best treatment option for patients with high-risk myelodysplastic syndromes (MDS). However, only half of treated patients respond and of these almost all eventually relapse. New treatment options are urgently needed to improve the clinical management of these patients. Here, we perform a loss-of-function shRNA screen and identify the histone acetyl transferase and transcriptional co-activator, CREB binding protein (CBP), as a major regulator of AZA sensitivity. Compounds inhibiting the activity of CBP and the closely related p300 synergistically reduce viability of MDS-derived AML cell lines when combined with AZA. Importantly, this effect is specific for the RNA-dependent functions of AZA and not observed with the related compound decitabine that is only incorporated into DNA. The identification of immediate target genes leads us to the unexpected finding that the effect of CBP/p300 inhibition is mediated by globally down regulating protein synthesis., Azacitidine (AZA) treatment is used for patients with myelodysplasias that cannot undergo bone marrow transplantation; however, AZA treatment is only partially effective. Here the authors show synergy of AZA with compounds inhibiting the chromatin regulators CBP and p300, which is mediated by the RNA-dependent functions of AZA affecting protein translation.
- Published
- 2021
16. Severe metabolic alterations in liver cancer lead to ERK pathway activation and drug resistance
- Author
-
Verodia Charlestin, Andreas Zimmer, Biljana Blagojevic, Peter Sajjakulnukit, Marina Pasca di Magliano, Nadia Battello, Norbert Gretz, Bedair Dewidar, Stefan Wölfl, Zeribe C. Nwosu, Matthew H. Ward, Jens U. Marquardt, Steven Dooley, Jonathan P. Sleeman, Ute Hofmann, S Pereira, Mei Han, Weronika Piorońska, Carolina De La Torre, Matthias P. Ebert, Pavlo Holenya, Costas A. Lyssiotis, D Castven, Julia Lochead, Christoph Meyer, and Li Zhang
- Subjects
Life sciences ,biology ,0301 basic medicine ,Sorafenib ,MAPK/ERK pathway ,Carcinoma, Hepatocellular ,Research paper ,MAP Kinase Signaling System ,Glutamine ,Proliferation ,lcsh:Medicine ,Antineoplastic Agents ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,ddc:570 ,medicine ,Serine ,Humans ,HCC ,Protein Kinase Inhibitors ,Cell Proliferation ,lcsh:R5-920 ,Cell growth ,Chemistry ,Kinase ,Microarray analysis techniques ,Liver Neoplasms ,lcsh:R ,General Medicine ,Hep G2 Cells ,digestive system diseases ,Metabolic pathway ,030104 developmental biology ,Anaerobic glycolysis ,Drug Resistance, Neoplasm ,Kinase inhibitors ,030220 oncology & carcinogenesis ,Cancer research ,Metabolome ,Metabolic state ,Aerobic glycolysis ,Transcriptome ,lcsh:Medicine (General) ,medicine.drug - Abstract
Background: The extracellular signal-regulated kinase (ERK) pathway regulates cell growth, and is hyper-activated and associated with drug resistance in hepatocellular carcinoma (HCC). Metabolic pathways are profoundly dysregulated in HCC. Whether an altered metabolic state is linked to activated ERK pathway and drug response in HCC is unaddressed. Methods: We deprived HCC cells of glutamine to induce metabolic alterations and performed various assays, including metabolomics (with 13C-glucose isotope tracing), microarray analysis, and cell proliferation assays. Glutamine-deprived cells were also treated with kinase inhibitors (e.g. Sorafenib, Erlotinib, U0126 amongst other MEK inhibitors). We performed bioinformatics analysis and stratification of HCC tumour microarrays to determine upregulated ERK gene signatures in patients. Findings: In a subset of HCC cells, the withdrawal of glutamine triggers a severe metabolic alteration and ERK phosphorylation (pERK). This is accompanied by resistance to the anti-proliferative effect of kinase inhibitors, despite pERK inhibition. High intracellular serine is a consistent feature of an altered metabolic state and contributes to pERK induction and the kinase inhibitor resistance. Blocking the ERK pathway facilitates cell proliferation by reprogramming metabolism, notably enhancing aerobic glycolysis. We have identified 24 highly expressed ERK gene signatures that their combined expression strongly indicates a dysregulated metabolic gene network in human HCC tissues. Interpretation: A severely compromised metabolism lead to ERK pathway induction, and primes some HCC cells to pro-survival phenotypes upon ERK pathway blockade. Our findings offer novel insights for understanding, predicting and overcoming drug resistance in liver cancer patients. Fund: DFG, BMBF and Sino-German Cooperation Project Keywords: Metabolic state, Kinase inhibitors, Glutamine, Serine, Proliferation, Aerobic glycolysis, HCC
- Published
- 2020
17. A complementary study approach unravels novel players in the pathoetiology of Hirschsprung disease
- Author
-
Melanie Bewerunge-Hudler, Beate Niesler, Thomas Thumberger, Isabella Ceccherini, Marta Rusmini, Joseph Tilghman, Phillipp Romero, Leonie Carstensen, Ana Torroglosa, Ralph Röth, Erwin Brosens, Patrick Günther, Norbert Gretz, Christian P. Schaaf, Berta Luzón-Toro, Robert M.W. Hofstra, Volker Eckstein, Carolina De La Torre, Jutta Scheuerer, Stefanie Schmitteckert, Jill A. Rosenfeld, Julia Volz, Felix Lasitschka, Duco Schriemer, Merce Garcia-Barcelo, Cornelia Thöni, Clara S. Tang, Tanja Mederer, Cristina Martínez, Paul K.H. Tam, Gudrun A. Rappold, Stefan Holland-Cunz, Nagarajan Paramasivam, Salud Borrego, Heidelberg Stiftung Chirurgie, Heinz and Heide Dürr Stiftung, Heidelberg University Hospital, Heidelberg Biosciences International Graduate School, German Academic Scholarship Foundation, Instituto de Salud Carlos III, Ministerio de Ciencia, Innovación y Universidades (España), European Commission, and Clinical Genetics
- Subjects
Male ,Cancer Research ,Candidate gene ,Cellular differentiation ,Gene Expression ,PROTEIN ,QH426-470 ,Pediatrics ,ATP Binding Cassette Transporter, Subfamily D, Member 1 ,Transcriptome ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Medicine and Health Sciences ,BRAIN ,Genetics (clinical) ,Exome sequencing ,RISK ,0303 health sciences ,Computer-Aided Drug Design ,Neural crest ,Cell Differentiation ,Protein Inhibitors of Activated STAT ,TRANSCRIPTION FACTORS ,Pediatric Gastroenterology ,Gene Cloning ,Sterol Regulatory Element Binding Protein 1 ,Neuronal Differentiation ,Research Article ,Drug Research and Development ,GENES ,Cell Survival ,MIGRATION ,In silico ,MODELS ,ADRENOLEUKODYSTROPHY ,Gastroenterology and Hepatology ,Computational biology ,Biology ,Research and Analysis Methods ,Cell Line ,03 medical and health sciences ,Exome Sequencing ,NERVOUS-SYSTEM DEVELOPMENT ,Genetics ,ATP7A ,Animals ,Humans ,Computer Simulation ,Hirschsprung Disease ,Molecular Biology Techniques ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Cell Proliferation ,030304 developmental biology ,Pharmacology ,Gene Expression Profiling ,Biology and Life Sciences ,Infant ,Human Genetics ,Human genetics ,Gene expression profiling ,Disease Models, Animal ,Copper-Transporting ATPases ,Drug Design ,030217 neurology & neurosurgery ,Cloning ,Developmental Biology - Abstract
[Abstract] Hirschsprung disease (HSCR, OMIM 142623) involves congenital intestinal obstruction caused by dysfunction of neural crest cells and their progeny during enteric nervous system (ENS) development. HSCR is a multifactorial disorder; pathogenetic variants accounting for disease phenotype are identified only in a minority of cases, and the identification of novel disease-relevant genes remains challenging. In order to identify and to validate a potential disease-causing relevance of novel HSCR candidate genes, we established a complementary study approach, combining whole exome sequencing (WES) with transcriptome analysis of murine embryonic ENS-related tissues, literature and database searches, in silico network analyses, and functional readouts using candidate gene-specific genome-edited cell clones. WES datasets of two patients with HSCR and their non-affected parents were analysed, and four novel HSCR candidate genes could be identified: ATP7A, SREBF1, ABCD1 and PIAS2. Further rare variants in these genes were identified in additional HSCR patients, suggesting disease relevance. Transcriptomics revealed that these genes are expressed in embryonic and fetal gastrointestinal tissues. Knockout of these genes in neuronal cells demonstrated impaired cell differentiation, proliferation and/or survival. Our approach identified and validated candidate HSCR genes and provided further insight into the underlying pathomechanisms of HSCR., [Author summary] Hirschsprung disease (HSCR) is a rare developmental disorder. It leads to the absence of enteric nerve cells (aganglionosis) in the large intestine and is caused by functional defects of neuronal precursor cells during embryonic development of the gut nervous system. The aganglionosis manifests as a variety of symptoms including impaired peristalsis and the formation of a pathogenic dilatation of the intestine (megacolon). The etiology of HSCR is considered to be multifactorial. Variants in more than 20 genes have been reported to be overrepresented in HSCR and replicated in independent cohorts. However, variants in those risk genes account for only 30% of all cases, suggesting that many more genes have to be implicated in the development of HSCR. As the identification and the subsequent validation of novel gene variants to be disease-causing or not, still remains a major challenge, we established and applied a complementary study pipeline. This enabled us to identify four novel candidate genes in two HSCR patients and to validate their potential disease relevance. Our approach represents a suitable way to dissect the complex genetic architecture underlying HSCR., This study was supported by the Heidelberg Stiftung Chirurgie (https://www.stiftung-chirurgie.de) (P. Romero and B. Niesler), the Heinz and Heide Dürr Stiftung (https://www.heinzundheideduerrstiftung.de) (B. Niesler and P. Romero; 2017/2.2.1/04), the Heidelberg University Hospital (https://www.heidelberg-university-hospital.com) (G. Rappold), and the Dres. Majic/Majic Schlez Stiftung (T. Mederer). Tanja Mederer is a PhD fellow of HBIGS (http://www.hbigs.uni-heidelberg.de) and was funded by the Studienstiftung des Deutschen Volkes (https://www.studienstiftung.de). Cristina Martínez is supported by Instituto de Salud Carlos III, Subdirección General de Investigación Sanitaria, Ministerio de Ciencia, Innovación y Universidades (https://www.isciii.es) (CP18/00116). Salud Borrego was supported by Instituto de Salud Carlos III (https://www.isciii.es) through the project "PI16/0142" and "PI19/01550" (Co-funded by European Regional Development Fund/European Social Fund "A way to make Europe"/"Investing in your future").
- Published
- 2020
18. miRNA profiling of biliary intraepithelial neoplasia reveals stepwise tumorigenesis in distal cholangiocarcinoma via the miR-451a/ATF2 axis
- Author
-
Benjamin Goeppert, Junfang Ji, Christian Rupp, Carsten Sticht, Stephanie Roessler, Jun Hu, Peter Schirmacher, Christoph Springfeld, Katrin Hoffmann, Carolina De La Torre, Jesus M. Banales, Thomas Albrecht, Yi Xiao, Bruno Köhler, Moritz Loeffler, Monika Nadja Vogel, Xiyang Wei, Anita Pathil-Warth, Martina Kirchner, and Arianeb Mehrabi
- Subjects
0301 basic medicine ,Male ,Microarray ,Carcinogenesis ,ADAM10 ,Down-Regulation ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Cholangiocarcinoma ,03 medical and health sciences ,chemistry.chemical_compound ,ADAM10 Protein ,0302 clinical medicine ,Bile Ducts, Extrahepatic ,Cell Movement ,microRNA ,medicine ,Biomarkers, Tumor ,Humans ,Bilin ,Laser capture microdissection ,Activating Transcription Factor 2 ,Gene Expression Profiling ,Membrane Proteins ,Cell migration ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,chemistry ,Bile Duct Neoplasms ,030220 oncology & carcinogenesis ,Case-Control Studies ,Biliary Intraepithelial Neoplasia ,Cancer research ,Female ,Amyloid Precursor Protein Secretases ,Carcinoma in Situ - Abstract
Distal cholangiocarcinoma (dCCA) is a biliary tract cancer with a dismal prognosis and is often preceded by biliary intraepithelial neoplasia (BilIN), representing the most common biliary non-invasive precursor lesion. BilIN are histologically well defined but have not so far been characterised systematically at the molecular level. The aim of this study was to determine miRNA-regulated genes in cholangiocarcinogenesis via BilIN. We used a clinicopathologically well-characterised cohort of 12 dCCA patients. Matched samples of non-neoplastic biliary epithelia, BilIN and invasive tumour epithelia of each patient were isolated from formalin-fixed paraffin-embedded tissue sections by laser microdissection. The resulting 36 samples were subjected to total RNA extraction and the expression of 798 miRNAs was assessed using the Nanostring® technology. Candidate miRNAs were validated by RT-qPCR and functionally investigated following lentiviral overexpression in dCCA-derived cell lines. Potential direct miRNA target genes were identified by microarray and prediction algorithms and were confirmed by luciferase assay. We identified 49 deregulated miRNAs comparing non-neoplastic and tumour tissue. Clustering of these miRNAs corresponded to the three stages of cholangiocarcinogenesis, supporting the concept of BilIN as a tumour precursor. Two downregulated miRNAs, i.e. miR-451a (-10.9-fold down) and miR-144-3p (-6.3-fold down), stood out by relative decrease. Functional analyses of these candidates revealed a migration inhibitory effect in dCCA cell lines. Activating transcription factor 2 (ATF2) and A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) were identified as direct miR-451a target genes. Specific ATF2 inhibition by pooled siRNAs reproduced the inhibitory impact of miR-451a on cancer cell migration. Thus, our data support the concept of BilIN as a direct precursor of invasive dCCA at the molecular level. In addition, we identified miR-451a and miR-144-3p as putative tumour suppressors attenuating cell migration by inhibiting ATF2 in the process of dCCA tumorigenesis. © The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2019
19. NOTCH target gene HES5 mediates oncogenic and tumor suppressive functions in hepatocarcinogenesis
- Author
-
Stephan Singer, Matthias Schlesner, Matthias Bieg, Arianeb Mehrabi, Roland Eils, Jens U. Marquardt, Sarah Luiken, Gregor Warsow, Carsten Sticht, Carolin Ploeger, Thomas Longerich, Raisatun Sugiyanto, Carolina De La Torre, Benjamin Goeppert, Stephanie Roessler, Stefan Pusch, Peter Schirmacher, Norbert Gretz, and Angelika Fraas
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Carcinoma, Hepatocellular ,DNA Mutational Analysis ,HES5 ,Notch signaling pathway ,Biology ,medicine.disease_cause ,Article ,Cohort Studies ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Mutant protein ,Cell Movement ,Cell Line, Tumor ,Exome Sequencing ,Genetics ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,ddc:610 ,HES1 ,Molecular Biology ,Protein kinase B ,Cancer genetics ,Regulation of gene expression ,Mutation ,Receptors, Notch ,Liver Neoplasms ,Oncogenes ,Xenograft Model Antitumor Assays ,3. Good health ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,030104 developmental biology ,Liver ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Carcinogenesis ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
NOTCH receptor signaling plays a pivotal role in liver homeostasis and hepatocarcinogenesis. However, the role of NOTCH pathway mutations and the NOTCH target gene HES5 in liver tumorigenesis are poorly understood. Here we performed whole-exome sequencing of 54 human HCC specimens and compared the prevalence of NOTCH pathway component mutations with the TCGA-LIHC cohort (N = 364). In addition, we functionally characterized the NOTCH target HES5 and the patient-derived HES5-R31G mutation in vitro and in an orthotopic mouse model applying different oncogenic backgrounds, to dissect the role of HES5 in different tumor subgroups in vivo. We identified nonsynonymous mutations in 14 immediate NOTCH pathway genes affecting 24.1% and 16.8% of HCC patients in the two independent cohorts, respectively. Among these, the HES5-R31G mutation was predicted in silico to have high biological relevance. Functional analyses in cell culture showed that HES5 reduced cell migration and clonogenicity. Further analyses revealed that the patient-derived HES5-R31G mutant protein was non-functional due to loss of DNA binding and greatly reduced nuclear localization. Furthermore, HES5 exhibited a negative feedback loop by directly inhibiting the NOTCH target HES1 and downregulated the pro-proliferative MYC targets ODC1 and LDHA. Interestingly, HES5 inhibited MYC-dependent hepatocarcinogenesis, whereas it promoted AKT-dependent liver tumor formation and stem cell features in a murine model. Thus, NOTCH pathway component mutations are commonly observed in HCC. Furthermore, the NOTCH target gene HES5 has both pro- and anti-tumorigenic functions in liver cancer proposing a driver gene dependency and it promotes tumorigenesis with its interaction partner AKT.
- Published
- 2019
20. Inhibition of miR30a-3p by sulforaphane enhances gap junction intercellular communication in pancreatic cancer
- Author
-
Ingrid Herr, Michael Schäfer, Carsten Sticht, Libo Yin, Wolfgang Gross, Jury Gladkich, Carolina De La Torre, Norbert Gretz, Xi Xiao, Christina Georgikou, and Svetlana Karakhanova
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Connexin ,In situ hybridization ,Cell Communication ,Deoxycytidine ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Isothiocyanates ,Pancreatic cancer ,Cell Line, Tumor ,microRNA ,medicine ,Animals ,Humans ,In Situ Hybridization ,Aged ,Gap Junctions ,Transfection ,Middle Aged ,medicine.disease ,Gemcitabine ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,MicroRNAs ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Connexin 43 ,Sulfoxides ,Isothiocyanate ,Cancer research ,Heterografts ,Female ,medicine.drug ,Sulforaphane ,Signal Transduction - Abstract
The therapy resistance of pancreatic cancer is associated with the loss of gap junction intercellular communication and connexin 43 expression. The broccoli isothiocyanate sulforaphane restored these features and therapy sensitivity. We investigated whether microRNA signaling is involved. Established cell lines and a patient tissue array (n = 96) were evaluated by miRNA and gene array, bioinformatics, expression studies, in situ hybridization and immunohistochemistry. Sulforaphane inhibited the expression of our top candidate miR30a-3p. Upon transfection of miR30a-3p inhibitors, the gemcitabine bystander effect, Cx43 expression and intercellular communication increased, whereas miR30a-3p mimics inhibited the luciferase activity of a Cx43 3′-UTR construct. miR30a-3p-overexpressing tumor xenografts had a decreased tumor volume and increased gemcitabine sensitivity. In patient tissues, a higher expression of miR30a-3p and a lower expression of Cx43 correlated with malignancy. These findings provide new knowledge and suggest sulforaphane as cotreatment for pancreatic cancer.
- Published
- 2019
21. Abstract 5416: Positioning the NQO1-bioactivatable drug isobutyl-deoxynyboquinone in diffuse intrinsic pontine glioma (DIPG): an exceptional therapeutic opportunity in pediatric brain tumor
- Author
-
Carolina De La Torre, Pere Llinas Arias, Paul J. Hergenrother, Manel Esteller, Andres Morales La Madrid, Vanessa Ortiz Barahona, Maxime Janin, and Angel M. Carcaboso
- Subjects
Cancer Research ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,Pediatric cancer ,Radiation therapy ,Efficacy ,Oncology ,Cell culture ,In vivo ,Apoptosis ,medicine ,Transcriptional regulation ,Cancer research ,business - Abstract
Purpose: Diffuse Intrinsic Pontine Glioma (DIPG), is a very aggressive pediatric cancer with poor overall survival and no effective treatment. Despite numerous clinical trials, the overall survival remains at 9 months after diagnosis and only radiotherapy has shown a relative efficacy. Therefore, it is primordial to increase our understanding of the biology of DIPG tumors, as well as finding new healthcare strategies to tackle this pediatric disease. Recently, we discovered that NQO1, a stress-related protein, was overexpressed in 3 out of 6 DIPG patient-derived cell lines, as well as in half of the primary tissues. Interestingly, NQO1 overexpression can be targeted by substrates that induce an excessive oxidative stress into the cells that finally push them to apoptosis, while normal surrounding tissues with basal expression of NQO1 would be keep safe. Experimental procedure: This discovery prompted us to test in in vitro and in vivo assays a promising drug named isobutyl-deoxynyboquinone (IB-DNQ): a NQO1 bioactivatable substrate. We first tested the drug response of the 6 cell lines before testing it on orthotopic xenograft mouse models. We also investigated by which mechanism NQO1 is regulated in DIPG. Finally, we performed experiments in order to decipher by which mechanism NQO1 is regulated in DIPG. Results: We saw that NQO1 overexpressing cells are very sensitive to the drug, compared to the cell lines with normal expression. Moreover, we started to validate the use of IB-DNQ in vivo using a DIPG cell line treated or not (mock) with IB-DNQ and also a NQO1-knockdown model in orthotopic xenograft mice models, confirming that IB-DNQ crosses the blood brain barrier and increases the overall survival. NQO1 increased expression in DIPG is surprisingly not due to a NRF2-mediated transcriptional regulation as we did not observe a correlation between NQO1 transcript abundance and protein expression in DIPG cell lines, indicating that NRF2 pathway could not be implicated, but translational or post-translational regulation could be operating. We performed a polysome profiling of 4 patient-derived cell lines, a protein stability assay and a large scale proteome for the 6 cell lines (pending results). Conclusion: Our preliminary results, including drug efficacy, are very encouraging for the development of this new therapeutic in DIPG. This discovery represents a promising opportunity to tackle this devastating disease and a new hope for the patients and their families. Citation Format: Maxime Henri Janin, Vanessa Ortiz Barahona, Pere Llinas Arias, Carolina De La Torre, Angel Montero Carcaboso, Andres Morales La Madrid, Paul Hergenrother, Manel Esteller. Positioning the NQO1-bioactivatable drug isobutyl-deoxynyboquinone in diffuse intrinsic pontine glioma (DIPG): an exceptional therapeutic opportunity in pediatric brain tumor [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5416.
- Published
- 2020
- Full Text
- View/download PDF
22. SOX9 expression decreases survival of patients with intrahepatic cholangiocarcinoma by conferring chemoresistance
- Author
-
Cédric Coulouarn, Carolina De La Torre, Damien Bergeat, Honglei Weng, Roman Liebe, Jun Li, Steven Dooley, Matthias P. Ebert, Laurent Sulpice, Tao Lin, Norbert Gretz, X Yuan, Heidelberg University, University of Tübingen, Nutrition, Métabolismes et Cancer (NuMeCan), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES), University of Heidelberg, Medical Faculty, Saarland University [Saarbrücken], Chinese-German Cooperation Group project [GZ 1263], Deutsche Forschungsgemeinschaft [Do373/13-1], Ligue Contre le Cancer - Comite departemental des Cotes-d'Armor, Novartis Oncology, Institut National de la Recherche Agronomique (INRA)-Université de Rennes (UR)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Jonchère, Laurent
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system ,Cell cycle checkpoint ,animal structures ,Microarray ,[SDV]Life Sciences [q-bio] ,Antineoplastic Agents ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Deoxycytidine ,Article ,Cholangiocarcinoma ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,stomatognathic system ,Cancer stem cell ,Biomarkers, Tumor ,Chemotherapy ,Humans ,Medicine ,Gene silencing ,CHEK1 ,Cell Proliferation ,Gene knockdown ,business.industry ,Liver Diseases ,Bile duct cancer ,SOX9 Transcription Factor ,Gene signature ,musculoskeletal system ,Survival Analysis ,Gemcitabine ,3. Good health ,[SDV] Life Sciences [q-bio] ,Treatment Outcome ,030104 developmental biology ,Bile Duct Neoplasms ,Oncology ,Drug Resistance, Neoplasm ,Chronic Disease ,embryonic structures ,Cancer research ,business ,medicine.drug - Abstract
International audience; BACKGROUND:Sex-determining region Y-box (SRY-box) containing gene 9 (SOX9) expression confers cancer stem cell features. However, SOX9 function in intrahepatic cholangiocarcinoma (iCCA) is unknown. This study investigated the effects and underlying mechanisms of SOX9 in iCCA.METHODS:SOX9 expression in 59 iCCA patients was examined by immunohistochemistry. The association between SOX9 expression and clinical outcome was evaluated. Gene signature and biological functions of SOX9 in iCCA were examined in vitro.RESULTS:iCCA patients with high SOX9 expression had shorter survival time than those with low SOX9. In patients receiving chemotherapy, median survival time in patients with low and high levels of SOX9 were 62 and 22 months, respectively. In vitro, gemcitabine increased SOX9 expression in iCCA cells. When SOX9 was knocked down, gemcitabine-induced apoptosis was markedly increased. Silencing SOX9 significantly inhibited gemcitabine-induced phosphorylation of checkpoint kinase 1, a key cell cycle checkpoint protein that coordinates the DNA damage response and inhibited the expression of multidrug resistance genes. Microarray analyses showed that SOX9 knockdown in CCA cells altered gene signatures associated with multidrug resistance and p53 signalling.CONCLUSIONS:SOX9 governs the response of CCA cells to chemotherapy. SOX9 is a biomarker to select iCCA patients eligible for efficient chemotherapy.
- Published
- 2018
- Full Text
- View/download PDF
23. Epigenetically regulated chromosome 14q32 miRNA cluster induces metastasis and predicts poor prognosis in lung adenocarcinoma patients
- Author
-
Fabian J. Theis, Norbert Gretz, Steffen Sass, Marco Albrecht, Justo Lorenzo-Bermejo, Damian Stichel, Carolina De La Torre, Arne Warth, Francisco J. Sánchez-Rivera, Peter Schirmacher, Adriana Pitea, J Schmitt, Julian Gutekunst, Kai Breuhahn, Nikola S. Müller, Darjus F. Tschaharganeh, Margarita González-Vallinas, Michael Meister, Thomas Muley, Franziska Matthäus, Manuel Rodríguez-Paredes, and Carsten Sticht
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Bisulfite sequencing ,Adenocarcinoma of Lung ,Biology ,Metastasis ,Epigenesis, Genetic ,Cohort Studies ,03 medical and health sciences ,microRNA ,medicine ,Humans ,Neoplasm Metastasis ,Lung cancer ,Molecular Biology ,Lymph node ,Chromosomes, Human, Pair 14 ,Microarray analysis techniques ,miARN ,Cáncer ,medicine.disease ,Prognosis ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,DNA methylation ,Cancer research ,Adenocarcinoma ,Female ,3207.13 Oncología - Abstract
Producción Científica, Most lung cancer deaths are related to metastases, which indicates the necessity of detecting and inhibiting tumor cell dissemination. Here, we aimed to identify miRNAs involved in metastasis of lung adenocarcinoma as prognostic biomarkers and therapeutic targets. To that end, lymph node metastasis– associated miRNAs were identified in The Cancer Genome Atlas lung adenocarcinoma patient cohort (sequencing data; n ¼ 449) and subsequently validated by qRT-PCR in an independent clinical cohort (n ¼ 108). Overexpression of miRNAs located on chromosome 14q32 was associated with metastasis in lung adenocarcinoma patients. Importantly, Kaplan–Meier analysis and log-rank test revealed that higher expression levels of individual 14q32 miRNAs (mir-539, mir-323b, and mir487a) associated with worse disease-free survival of never-smoker patients. Epigenetic analysis including DNA methylation microarray data and bisulfite sequencing validation demonstrated that the induction of 14q32 cluster correlated with genomic hypomethylation of the 14q32 locus. CRISPR activation technology, applied for the first time to functionally study the increase of clustered miRNA levels in a coordinated manner, showed that simultaneous overexpression of 14q32 miRNAs promoted tumor cell migratory and invasive properties. Analysis of individual miRNAs by mimic transfection further illustrated that miR-323b-3p, miR-487a-3p, and miR-539-5p significantly contributed to the invasive phenotype through the indirect regulation of different target genes. In conclusion, overexpression of 14q32 miRNAs, associated with the respective genomic hypomethylation, promotes metastasis and correlates with poor patient prognosis in lung adenocarcinoma
- Published
- 2018
- Full Text
- View/download PDF
24. KAT6B is a tumor suppressor histone H3 lysine 23 acetyltransferase undergoing genomic loss in small cell lung cancer
- Author
-
Alberto Villanueva, Fernando Setien, Alejandro Vaquero, Ugo Pastorino, Reika Iwakawa, Jun Yokota, August Vidal, Katalin Szakszon, Carolina De La Torre, Luca Roz, María Berdasco, Sebastian Moran, Conceição S. Moura, Guntram Borck, Enrique Vidal, Catia Moutinho, Fátima Carneiro, Laia Simó-Riudalbas, Ilse Zondervan, Marta Soler, Suvi Savola, Manel Esteller, Silvia Barceló-Batllori, Montserrat Pérez-Salvia, Holger Heyn, Takashi Kohno, Antonio Gomez, and Anna Martínez-Cardús
- Subjects
Cancer Research ,Chromatin Immunoprecipitation ,Lung Neoplasms ,Mice, Nude ,Irinotecan ,Histones ,Histone H3 ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Epigenetics ,Cancer epigenetics ,RNA, Messenger ,RNA, Neoplasm ,Carcinoma, Small Cell ,Neoplasm Metastasis ,RNA, Small Interfering ,Antineoplastic Agents, Alkylating ,Histone Acetyltransferases ,biology ,Gene Expression Profiling ,Cancer ,Acetylation ,Histone acetyltransferase ,medicine.disease ,Molecular biology ,Neoplasm Proteins ,Histone ,Oncology ,Drug Resistance, Neoplasm ,Acetyltransferase ,biology.protein ,Heterografts ,Camptothecin ,RNA Interference ,Chromatin immunoprecipitation ,Protein Processing, Post-Translational ,Gene Deletion - Abstract
Recent efforts to sequence human cancer genomes have highlighted that point mutations in genes involved in the epigenetic setting occur in tumor cells. Small cell lung cancer (SCLC) is an aggressive tumor with poor prognosis, where little is known about the genetic events related to its development. Herein, we have identified the presence of homozygous deletions of the candidate histone acetyltransferase KAT6B, and the loss of the corresponding transcript, in SCLC cell lines and primary tumors. Furthermore, we show, in vitro and in vivo, that the depletion of KAT6B expression enhances cancer growth, while its restoration induces tumor suppressor–like features. Most importantly, we demonstrate that KAT6B exerts its tumor-inhibitory role through a newly defined type of histone H3 Lys23 acetyltransferase activity. Cancer Res; 75(18); 3936–45. ©2015 AACR.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.