29 results on '"Christina Stangl"'
Search Results
2. Partner independent fusion gene detection by multiplexed CRISPR-Cas9 enrichment and long read nanopore sequencing
- Author
-
Christina Stangl, Sam de Blank, Ivo Renkens, Liset Westera, Tamara Verbeek, Jose Espejo Valle-Inclan, Rocio Chamorro González, Anton G. Henssen, Markus J. van Roosmalen, Ronald W. Stam, Emile E. Voest, Wigard P. Kloosterman, Gijs van Haaften, and Glen R. Monroe
- Subjects
Science - Abstract
Fusion genes are a hallmarks of cancer, though breakpoint-position promiscuity restricts diagnostic detection. Here, the authors present FUDGE, a CRISPR-Cas9-based enrichment strategy for nanopore sequencing to identify target fusions irrespective of genomic breakpoint or fusion partner.
- Published
- 2020
- Full Text
- View/download PDF
3. Response-predictive gene expression profiling of glioma progenitor cells in vitro.
- Author
-
Sylvia Moeckel, Katharina Meyer, Petra Leukel, Fabian Heudorfer, Corinna Seliger, Christina Stangl, Ulrich Bogdahn, Martin Proescholdt, Alexander Brawanski, Arabel Vollmann-Zwerenz, Markus J Riemenschneider, Anja-Katrin Bosserhoff, Rainer Spang, and Peter Hau
- Subjects
Medicine ,Science - Abstract
BACKGROUND:High-grade gliomas are amongst the most deadly human tumors. Treatment results are disappointing. Still, in several trials around 20% of patients respond to therapy. To date, diagnostic strategies to identify patients that will profit from a specific therapy do not exist. METHODS:In this study, we used serum-free short-term treated in vitro cell cultures to predict treatment response in vitro. This approach allowed us (a) to enrich specimens for brain tumor initiating cells and (b) to confront cells with a therapeutic agent before expression profiling. RESULTS:As a proof of principle we analyzed gene expression in 18 short-term serum-free cultures of high-grade gliomas enhanced for brain tumor initiating cells (BTIC) before and after in vitro treatment with the tyrosine kinase inhibitor Sunitinib. Profiles from treated progenitor cells allowed to predict therapy-induced impairment of proliferation in vitro. CONCLUSION:For the tyrosine kinase inhibitor Sunitinib used in this dataset, the approach revealed additional predictive information in comparison to the evaluation of classical signaling analysis.
- Published
- 2014
- Full Text
- View/download PDF
4. CRISPR‐Cas9 enrichment, a new strategy in microbial metagenomics to investigate complex genomic regions: The case of an environmental integron
- Author
-
Eva Sandoval‐Quintana, Christina Stangl, Lionel Huang, Ivo Renkens, Robert Duran, Gijs van Haaften, Glen Monroe, Béatrice Lauga, and Christine Cagnon
- Subjects
Genetics ,Ecology, Evolution, Behavior and Systematics ,Biotechnology - Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Table 3 from Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
W.P. Kloosterman, Hugo J.G. Snippert, Emile E. Voest, Marco J. Koudijs, Robert M. van Es, Harmjan R. Vos, Ingrid Verlaan-Klink, Nizar Hami, Markus J. van Roosmalen, Jasmin B. Post, and Christina Stangl
- Abstract
Supplementary Table 3. Z-scores of significantly differential expressed genes (provided separately due to size)
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table 2 from Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
W.P. Kloosterman, Hugo J.G. Snippert, Emile E. Voest, Marco J. Koudijs, Robert M. van Es, Harmjan R. Vos, Ingrid Verlaan-Klink, Nizar Hami, Markus J. van Roosmalen, Jasmin B. Post, and Christina Stangl
- Abstract
Supplementary Table 2. Differential Gene Expression Analysis on BRAF (fusion) gene expressing organoid lines (provided separately due to size)
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
W.P. Kloosterman, Hugo J.G. Snippert, Emile E. Voest, Marco J. Koudijs, Robert M. van Es, Harmjan R. Vos, Ingrid Verlaan-Klink, Nizar Hami, Markus J. van Roosmalen, Jasmin B. Post, and Christina Stangl
- Abstract
Fusion genes can be oncogenic drivers in a variety of cancer types and represent potential targets for targeted therapy. The BRAF gene is frequently involved in oncogenic gene fusions, with fusion frequencies of 0.2%–3% throughout different cancers. However, BRAF fusions rarely occur in the same gene configuration, potentially challenging personalized therapy design. In particular, the impact of the wide variety of fusion partners on the oncogenic role of BRAF during tumor growth and drug response is unknown. Here, we used patient-derived colorectal cancer organoids to functionally characterize and cross-compare BRAF fusions containing various partner genes (AGAP3, DLG1, and TRIM24) with respect to cellular behavior, downstream signaling activation, and response to targeted therapies. We demonstrate that 5′ fusion partners mainly promote canonical oncogenic BRAF activity by replacing the auto-inhibitory N-terminal region. In addition, the 5′ partner of BRAF fusions influences their subcellular localization and intracellular signaling capacity, revealing distinct subsets of affected signaling pathways and altered gene expression. Presence of the different BRAF fusions resulted in varying sensitivities to combinatorial inhibition of MEK and the EGF receptor family. However, all BRAF fusions conveyed resistance to targeted monotherapy against the EGF receptor family, suggesting that BRAF fusions should be screened alongside other MAPK pathway alterations to identify patients with metastatic colorectal cancer to exclude from anti-EGFR–targeted treatment.Implications:Although intracellular signaling and sensitivity to targeted therapies of BRAF fusion genes are influenced by their 5′ fusion partner, we show that all investigated BRAF fusions confer resistance to clinically relevant EGFR inhibition.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure Legends from Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
W.P. Kloosterman, Hugo J.G. Snippert, Emile E. Voest, Marco J. Koudijs, Robert M. van Es, Harmjan R. Vos, Ingrid Verlaan-Klink, Nizar Hami, Markus J. van Roosmalen, Jasmin B. Post, and Christina Stangl
- Abstract
Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table 1 from Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
W.P. Kloosterman, Hugo J.G. Snippert, Emile E. Voest, Marco J. Koudijs, Robert M. van Es, Harmjan R. Vos, Ingrid Verlaan-Klink, Nizar Hami, Markus J. van Roosmalen, Jasmin B. Post, and Christina Stangl
- Abstract
Supplementary Table 1. KSEA kinase scores and kinase-substrate links identified in phosphoproteomic screen (provided separately due to size)
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table 4 from Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
W.P. Kloosterman, Hugo J.G. Snippert, Emile E. Voest, Marco J. Koudijs, Robert M. van Es, Harmjan R. Vos, Ingrid Verlaan-Klink, Nizar Hami, Markus J. van Roosmalen, Jasmin B. Post, and Christina Stangl
- Abstract
Supplementary Table 4. List of BRAF (fusion) gene and GFP-specific primers. Primer used for breakpoint PCR, HA-tag spanning PCR and for the gateway cloning
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figures 1-11 from Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
W.P. Kloosterman, Hugo J.G. Snippert, Emile E. Voest, Marco J. Koudijs, Robert M. van Es, Harmjan R. Vos, Ingrid Verlaan-Klink, Nizar Hami, Markus J. van Roosmalen, Jasmin B. Post, and Christina Stangl
- Abstract
S1. Impact of BRAF (fusion) gene expression on phosphorylation of AKT. S2. BRAF (fusion) gene mRNA and protein expression levels. S3. Phosphoproteomics screen in HEK293 cells reveals that BRAF fusions and BRAFV600E activate similar signaling pathways. S4. BRAF (fusion) gene expression and MAPK pathway activation in HEK293 cells. S5. BRAF (fusion) protein localization in HEK293 cells. S6. Establishment of optimal duration of dox induction for BRAF (fusion) protein expression. S7. Validation of phospho-proteins and transcriptomic analysis of BRAF (fusion) gene expressing P18T CRC organoid lines. S8. Expression of most differentially expressed genes in DLG1-BRAF expressing P18T organoids and CRC patients. S9. BRAF (fusion) genes confer resistance to EGFR inhibition. S10. Impact of afatinib exposure on pAKT levels in BRAF (fusion) gene expressing CRC organoids. S11. Differential sensitivities of BRAF (fusion) genes to combinatorial targeting of EGFR and MEK.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Materials and Methods from Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
W.P. Kloosterman, Hugo J.G. Snippert, Emile E. Voest, Marco J. Koudijs, Robert M. van Es, Harmjan R. Vos, Ingrid Verlaan-Klink, Nizar Hami, Markus J. van Roosmalen, Jasmin B. Post, and Christina Stangl
- Abstract
Supplementary Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table S1 from A Systematic Analysis of Oncogenic Gene Fusions in Primary Colon Cancer
- Author
-
Emile E. Voest, Jan N.M. Ijzermans, Marco J. Koudijs, Katharina Biermann, John A. Foekens, John W.M. Martens, Fried Zwartkruis, Armel Lefebvre, Marcel Smid, Anne van Galen, Salo Ooft, Zarina S. Lalmahomed, Ronne Brunekreef, Christina Stangl, Anieta M. Sieuwerts, Markus J. van Roosmalen, Mark Pieterse, Robert R.J. Coebergh van den Braak, and Wigard P. Kloosterman
- Abstract
Colon tumor sample overview, clinical characteristics, fusion genes and KRAS, NRAS, BRAF mutation status and sequencing statistics.
- Published
- 2023
- Full Text
- View/download PDF
14. Data from A Systematic Analysis of Oncogenic Gene Fusions in Primary Colon Cancer
- Author
-
Emile E. Voest, Jan N.M. Ijzermans, Marco J. Koudijs, Katharina Biermann, John A. Foekens, John W.M. Martens, Fried Zwartkruis, Armel Lefebvre, Marcel Smid, Anne van Galen, Salo Ooft, Zarina S. Lalmahomed, Ronne Brunekreef, Christina Stangl, Anieta M. Sieuwerts, Markus J. van Roosmalen, Mark Pieterse, Robert R.J. Coebergh van den Braak, and Wigard P. Kloosterman
- Abstract
Genomic rearrangements that give rise to oncogenic gene fusions can offer actionable targets for cancer therapy. Here we present a systematic analysis of oncogenic gene fusions among a clinically well-characterized, prospectively collected set of 278 primary colon cancers spanning diverse tumor stages and clinical outcomes. Gene fusions and somatic genetic variations were identified in fresh frozen clinical specimens by Illumina RNA-sequencing, the STAR fusion gene detection pipeline, and GATK RNA-seq variant calling. We considered gene fusions to be pathogenically relevant when recurrent, producing divergent gene expression (outlier analysis), or as functionally important (e.g., kinase fusions). Overall, 2.5% of all specimens were defined as harboring a relevant gene fusion (kinase fusions 1.8%). Novel configurations of BRAF, NTRK3, and RET gene fusions resulting from chromosomal translocations were identified. An R-spondin fusion was found in only one tumor (0.35%), much less than an earlier reported frequency of 10% in colorectal cancers. We also found a novel fusion involving USP9X-ERAS formed by chromothripsis and leading to high expression of ERAS, a constitutively active RAS protein normally expressed only in embryonic stem cells. This USP9X–ERAS fusion appeared highly oncogenic on the basis of its ability to activate AKT signaling. Oncogenic fusions were identified only in lymph node–negative tumors that lacked BRAF or KRAS mutations. In summary, we identified several novel oncogenic gene fusions in colorectal cancer that may drive malignant development and offer new targets for personalized therapy. Cancer Res; 77(14); 3814–22. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Data file from A Systematic Analysis of Oncogenic Gene Fusions in Primary Colon Cancer
- Author
-
Emile E. Voest, Jan N.M. Ijzermans, Marco J. Koudijs, Katharina Biermann, John A. Foekens, John W.M. Martens, Fried Zwartkruis, Armel Lefebvre, Marcel Smid, Anne van Galen, Salo Ooft, Zarina S. Lalmahomed, Ronne Brunekreef, Christina Stangl, Anieta M. Sieuwerts, Markus J. van Roosmalen, Mark Pieterse, Robert R.J. Coebergh van den Braak, and Wigard P. Kloosterman
- Abstract
Combined supplementary data file containing: -extended materials and methods, describing details of sample collection and isolation, sequencing data generation, cell culturing and protein analysis. -Supplementary Figure S1 showing patient inclusion in this study -Supplementary Figure S2 showing RT-PCR results of fusion gene verification -Supplementary Figure S3 showing Exonic and protein structure of three BRAF fusions -Supplementary Figure S4 showing the structure of an EML4-NTRK3 fusion gene and exonic expression of NTRK3 -Supplementary Figure S5 showing the structure and expression of a novel RRBP1-RET fusion -Supplementary Figure S6 showing the structure and expression of a novel USP9X-ERAS fusion gene. -Supplementary Figure S7 showing Rspondin fusion gene detection and expression -List of Three Supplementary Tables -Supplementary References
- Published
- 2023
- Full Text
- View/download PDF
16. Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity
- Author
-
Markus J. van Roosmalen, Jasmin B. Post, Wigard P. Kloosterman, Marco J. Koudijs, Emile E. Voest, Hugo J. Snippert, Christina Stangl, Nizar Hami, Harmjan R. Vos, Robert M. van Es, and Ingrid Verlaan-Klink
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system diseases ,Colorectal cancer ,medicine.medical_treatment ,HEK 293 cells ,Cancer ,Biology ,medicine.disease ,digestive system diseases ,TRIM24 ,Targeted therapy ,Fusion gene ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Oncogene Fusion ,Signal transduction ,neoplasms ,Molecular Biology - Abstract
Fusion genes can be oncogenic drivers in a variety of cancer types and represent potential targets for targeted therapy. The BRAF gene is frequently involved in oncogenic gene fusions, with fusion frequencies of 0.2%–3% throughout different cancers. However, BRAF fusions rarely occur in the same gene configuration, potentially challenging personalized therapy design. In particular, the impact of the wide variety of fusion partners on the oncogenic role of BRAF during tumor growth and drug response is unknown. Here, we used patient-derived colorectal cancer organoids to functionally characterize and cross-compare BRAF fusions containing various partner genes (AGAP3, DLG1, and TRIM24) with respect to cellular behavior, downstream signaling activation, and response to targeted therapies. We demonstrate that 5′ fusion partners mainly promote canonical oncogenic BRAF activity by replacing the auto-inhibitory N-terminal region. In addition, the 5′ partner of BRAF fusions influences their subcellular localization and intracellular signaling capacity, revealing distinct subsets of affected signaling pathways and altered gene expression. Presence of the different BRAF fusions resulted in varying sensitivities to combinatorial inhibition of MEK and the EGF receptor family. However, all BRAF fusions conveyed resistance to targeted monotherapy against the EGF receptor family, suggesting that BRAF fusions should be screened alongside other MAPK pathway alterations to identify patients with metastatic colorectal cancer to exclude from anti-EGFR–targeted treatment. Implications: Although intracellular signaling and sensitivity to targeted therapies of BRAF fusion genes are influenced by their 5′ fusion partner, we show that all investigated BRAF fusions confer resistance to clinically relevant EGFR inhibition.
- Published
- 2020
- Full Text
- View/download PDF
17. Optimizing Nanopore sequencing-based detection of structural variants enables individualized circulating tumor DNA-based disease monitoring in cancer patients
- Author
-
Chris J. de Witte, Roel Janssen, Wigard P. Kloosterman, Martijn P. Lolkema, Christina Stangl, Markus J. van Roosmalen, Jose Espejo Valle-Inclan, Anouk C. de Jong, Jean C. A. Helmijr, Sam de Blank, Maurice P.H.M. Jansen, Ivo Renkens, John W.M. Martens, Lisanne F. van Dessel, and Medical Oncology
- Subjects
Male ,Nanopore ,Cancer genome sequencing ,Method ,Genomics ,Computational biology ,QH426-470 ,Biology ,Real-Time Polymerase Chain Reaction ,Sensitivity and Specificity ,Circulating Tumor DNA ,Liquid biopsies ,Structural variation ,Prostate cancer ,SDG 3 - Good Health and Well-being ,Neoplasms ,Biomarkers, Tumor ,Genetics ,medicine ,Humans ,Digital polymerase chain reaction ,Molecular Biology ,Genetics (clinical) ,Cancer ,Liquid Biopsy ,Computational Biology ,Reproducibility of Results ,Sequence Analysis, DNA ,medicine.disease ,Human genetics ,Nanopore Sequencing ,Molecular Diagnostic Techniques ,Organ Specificity ,Genomic Structural Variation ,Medicine ,Molecular Medicine ,Female ,Nanopore sequencing - Abstract
Here, we describe a novel approach for rapid discovery of a set of tumor-specific genomic structural variants (SVs), based on a combination of low coverage cancer genome sequencing using Oxford Nanopore with an SV calling and filtering pipeline. We applied the method to tumor samples of high-grade ovarian and prostate cancer patients and validated on average ten somatic SVs per patient with breakpoint-spanning PCR mini-amplicons. These SVs could be quantified in ctDNA samples of patients with metastatic prostate cancer using a digital PCR assay. The results suggest that SV dynamics correlate with and may improve existing treatment-response biomarkers such as PSA.https://github.com/UMCUGenetics/SHARC.
- Published
- 2021
- Full Text
- View/download PDF
18. In vitro systematic drug testing reveals carboplatin, paclitaxel, and alpelisib as a potential novel combination treatment for adult granulosa cell tumors
- Author
-
S. T. Paijens, Hannah S. van Meurs, Elena Sendino Garví, Geertruida N. Jonges, Ronald P. Zweemer, Gijs van Haaften, J. W. Groeneweg, René H.M. Verheijen, Hans W. Nijman, Karen Duran, Ellen Stelloo, Christianne A.R. Lok, Petronella O. Witteveen, Christina Stangl, Joline F. Roze, Glen R. Monroe, Jurgen M.J. Piek, Ferdinando Sereno, Luc R.C.W. van Lonkhuijzen, Obstetrics and Gynaecology, CCA - Cancer Treatment and Quality of Life, CCA - Cancer biology and immunology, Translational Immunology Groningen (TRIGR), and Targeted Gynaecologic Oncology (TARGON)
- Subjects
0301 basic medicine ,Drug ,Cancer Research ,medicine.medical_treatment ,media_common.quotation_subject ,Cmax ,lcsh:RC254-282 ,Article ,Alpelisib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Ovarian cancer ,Targeted treatment ,medicine ,Viability assay ,Drug screens ,IC50 ,media_common ,Chemotherapy ,business.industry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Carboplatin ,030104 developmental biology ,Oncology ,chemistry ,Paclitaxel ,030220 oncology & carcinogenesis ,Granulosa cell tumors ,Cancer research ,business - Abstract
Adult granulosa cell tumors (AGCTs) arise from the estrogen-producing granulosa cells. Treatment of recurrence remains a clinical challenge, as systemic anti-hormonal treatment or chemotherapy is only effective in selected patients. We established a method to rapidly screen for drug responses in vitro using direct patient-derived cell lines in order to optimize treatment selection. The response to 11 monotherapies and 12 combination therapies, including chemotherapeutic, anti-hormonal, and targeted agents, were tested in 12 AGCT-patient-derived cell lines and an AGCT cell line (KGN). Drug screens were performed within 3 weeks after tissue collection by measurement of cell viability 72 h after drug application. The potential synergy of drug combinations was assessed. The human maximum drug plasma concentration (Cmax) and steady state (Css) thresholds obtained from available phase I/II clinical trials were used to predict potential toxicity in patients. Patient-derived AGCT cell lines demonstrated resistance to all monotherapies. All cell lines showed synergistic growth inhibition by combination treatment with carboplatin, paclitaxel, and alpelisib at a concentration needed to obtain 50% cell death (IC50) that are below the maximum achievable concentration in patients (IC50 <, Cmax). We show that AGCT cell lines can be rapidly established and used for patient-specific in vitro drug testing, which may guide treatment decisions. Combination treatment with carboplatin, paclitaxel, and alpelisib was consistently effective in AGCT cell lines and should be further studied as a potential effective combination for AGCT treatment in patients.
- Published
- 2021
19. A Systematic Analysis of Oncogenic Gene Fusions in Primary Colon Cancer
- Author
-
John W.M. Martens, Marcel Smid, Zarina S. Lalmahomed, Anieta M. Sieuwerts, Christina Stangl, Katharina Biermann, Anne van Galen, Armel Lefebvre, Jan N. M. IJzermans, Salo N. Ooft, Wigard P. Kloosterman, John A. Foekens, Ronne Brunekreef, Robert R. J. Coebergh van den Braak, Mark Pieterse, Emile E. Voest, Marco J. Koudijs, Fried J. T. Zwartkruis, Markus J. van Roosmalen, Surgery, Medical Oncology, and Pathology
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Carcinogenesis ,Colorectal cancer ,Somatic cell ,Biology ,medicine.disease_cause ,Fusion gene ,03 medical and health sciences ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Gene expression ,medicine ,Humans ,Oncogene Fusion ,Gene ,Aged ,Genetics ,Chromothripsis ,Gene Expression Profiling ,Middle Aged ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female ,KRAS ,Colorectal Neoplasms ,Signal Transduction - Abstract
Genomic rearrangements that give rise to oncogenic gene fusions can offer actionable targets for cancer therapy. Here we present a systematic analysis of oncogenic gene fusions among a clinically well-characterized, prospectively collected set of 278 primary colon cancers spanning diverse tumor stages and clinical outcomes. Gene fusions and somatic genetic variations were identified in fresh frozen clinical specimens by Illumina RNA-sequencing, the STAR fusion gene detection pipeline, and GATK RNA-seq variant calling. We considered gene fusions to be pathogenically relevant when recurrent, producing divergent gene expression (outlier analysis), or as functionally important (e.g., kinase fusions). Overall, 2.5% of all specimens were defined as harboring a relevant gene fusion (kinase fusions 1.8%). Novel configurations of BRAF, NTRK3, and RET gene fusions resulting from chromosomal translocations were identified. An R-spondin fusion was found in only one tumor (0.35%), much less than an earlier reported frequency of 10% in colorectal cancers. We also found a novel fusion involving USP9X-ERAS formed by chromothripsis and leading to high expression of ERAS, a constitutively active RAS protein normally expressed only in embryonic stem cells. This USP9X–ERAS fusion appeared highly oncogenic on the basis of its ability to activate AKT signaling. Oncogenic fusions were identified only in lymph node–negative tumors that lacked BRAF or KRAS mutations. In summary, we identified several novel oncogenic gene fusions in colorectal cancer that may drive malignant development and offer new targets for personalized therapy. Cancer Res; 77(14); 3814–22. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
20. Relief from detrimental consequences of chronic psychosocial stress in mice deficient for the metabotropic glutamate receptor subtype 7
- Author
-
Daniel Peterlik, Robert Strasser, Peter J. Flor, Dominik Grabski, Christina Stangl, Dominic Schmidt, Anna Bludau, and Nicole Uschold-Schmidt
- Subjects
Male ,0301 basic medicine ,Inflammation ,Anxiety ,Receptors, Metabotropic Glutamate ,Bioinformatics ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Adrenocorticotropic Hormone ,AMN082 ,Downregulation and upregulation ,medicine ,Animals ,Prefrontal cortex ,Mice, Knockout ,Pharmacology ,Body Weight ,Stressor ,Blockade ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Metabotropic glutamate receptor ,medicine.symptom ,Corticosterone ,Psychology ,Neuroscience ,Psychosocial ,Stress, Psychological ,030217 neurology & neurosurgery - Abstract
Chronic stress-related psychiatric conditions and comorbid somatic pathologies are an enormous public health concern in modern society. The etiology of these disorders is complex, with stressors holding a chronic and psychosocial component representing the most acknowledged risk factor. During the last decades, research on the metabotropic glutamate receptor (mGlu) system advanced dramatically and much attention was given to the role of the metabotropic glutamate receptor subtype 7 (mGlu7) in acute stress-related behavior and physiology. However, virtually nothing is known about the potential involvement of mGlu7 in chronic psychosocial stress-related conditions. Using the chronic subordinate colony housing (CSC, 19 days) in male mice, we addressed whether central mGlu7 is altered upon chronic psychosocial stressor exposure and whether genetic ablation of mGlu7 interferes with the multitude of chronic stress-induced alterations. CSC exposure resulted in a downregulation of mGlu7 mRNA transcript levels in the prefrontal cortex, a brain region relevant for stress-related behaviors and physiology. Interestingly, mGlu7 deficiency relieved multiple chronic stress-induced alterations including the CSC-induced anxiety-prone phenotype; mGlu7 ablation also ameliorated CSC-induced physiological and immunological consequences such as hypothalamo-pituitary-adrenal (HPA) axis dysfunctions and colonic inflammation, respectively. Together, our findings provide first evidence for the involvement of mGlu7 in a wide range of behavioral and physiological alterations in response to chronic psychosocial stressor exposure. Moreover, the stress-protective phenotype of genetic mGlu7 ablation suggests mGlu7 pharmacological blockade to be a relevant option for the treatment of chronic stress-related emotional and somatic dysfunctions. This article is part of the Special Issue entitled ‘Metabotropic Glutamate Receptors, 5 years on’.
- Published
- 2017
- Full Text
- View/download PDF
21. Type-7 metabotropic glutamate receptors negatively regulate α1-adrenergic receptor signalling
- Author
-
Ferdinando Nicoletti, Luisa Iacovelli, Christina Stangl, Giuseppe Battaglia, Peter J. Flor, Antonio De Blasi, Rosamaria Orlando, Luisa Di Menna, Daniel Peterlik, Nicole Uschold-Schmidt, Valeria Bruno, and Gemma Molinaro
- Subjects
0301 basic medicine ,Pharmacology ,Agonist ,medicine.medical_specialty ,G protein-coupled receptor kinase ,medicine.drug_class ,Chemistry ,Stimulation ,Receptor antagonist ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,030104 developmental biology ,0302 clinical medicine ,Metabotropic receptor ,Endocrinology ,Metabotropic glutamate receptor ,Internal medicine ,medicine ,Receptor ,030217 neurology & neurosurgery ,G protein-coupled receptor - Abstract
We studied the interaction between mGlu7 and alpha(1)-adrenergic receptors in heterologous expression systems, brain slices, and living animals. L-2-Amino-4-phosphonobutanoate (L-AP4), and L-serine-O-phosphate (L-SOP), which activate group III mGlu receptors, restrained the stimulation of polyphosphoinositide (PI) hydrolysis induced by the alpha(1)-adrenergic receptor agonist, phenylephrine, in HEK 293 cells co-expressing alpha(1)-adrenergic and mGlu7 receptors. The inibitory action of L-AP4 was abrogated by (i) the mGlu7 receptor antagonist, XAP044; (ii) the C-terminal portion of type-2 G protein coupled receptor kinase; and (iii) the MAP kinase inhibitors, U0126 and PD98059. This suggests that the functional interaction between mGlu7 and alpha(1)-adrenergic receptors was mediated by the beta gamma-subunits of the G(i) protein and required the activation of the MAP kinase pathway. Remarkably, activation of neither mGlu2 nor mGlu4 receptors reduced alpha(1)-adrenergic receptor-mediated PI hydrolysis. In mouse cortical slices, both L-AP4 and L-SOP were able to attenuate norepinephrine- and phenylephrine-stimulated PI hydrolysis at concentrations consistent with the activation of mGlu7 receptors. L-AP4 failed to affect norepinephrine-stimulated PI hydrolysis in cortical slices from mGlu7(-/-) mice, but retained its inhibitory activity in slices from mGlu4(-/-) mice. At behavioural level, i.c.v. injection of phenylephrine produced antidepressant-like effects in the forced swim test. The action of phenylephrine was attenuated by L-SOP, which was inactive per se. Finally, both phenylephrine and L-SOP increased corticosterone levels in mice, but the increase was halved when the two drugs were administered in combination. Our data demonstrate that alpha(1)-adrenergic and mGlu7 receptors functionally interact and suggest that this interaction might be targeted in the treatment of stress-related disorders. (C) 2016 Elsevier Ltd. All rights reserved.
- Published
- 2017
- Full Text
- View/download PDF
22. Cancer modeling in colorectal organoids reveals intrinsic differences between oncogenic RAS and BRAF variants
- Author
-
Jasmin B. Post, Nizar Hami, Lohuis J, Christina Stangl, Ellen Stelloo, van de Ven M, Hjg Snippert, van Rheenen J, de Korte-Grimmerink R, and Verlaan I
- Subjects
MAPK/ERK pathway ,0303 health sciences ,Oncogene ,Mutant ,Cancer ,Biology ,medicine.disease ,Phenotype ,digestive system diseases ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Organoid ,CRISPR ,neoplasms ,030304 developmental biology - Abstract
Colorectal cancers (CRCs) with oncogenic mutations in RAS and BRAF are associated with anti-EGFR therapy resistance. Consequently, all RAS mutant CRC patients are being excluded from this therapy. However, heterogeneity in drug response has been reported between RAS mutant CRC patients. It is poorly understood to what extent such differences are derived from different genetic backgrounds or intrinsic differences between the various RAS pathway mutations. Therefore, using CRISPR technology we generated an isogenic panel of patient-derived CRC organoids with various RAS pathway mutations (i.e. KRASG12D, BRAFV600E, KRASG13D and NRASG12D). All RAS pathway mutants promote ERK activation and tumor growth. However, KRASG12D and BRAFV600E mutations in particular conferred robust resistance to anti-EGFR therapy, both in vitro and in vivo. Moreover, untreated KRASG13D mutants showed fastest growth in mice but remained sensitive to anti-EGFR therapy. Together, introducing mutation-specific oncogene signaling in CRC organoids resembles clinical phenotypes and improves understanding of genotype-phenotype correlations.
- Published
- 2019
- Full Text
- View/download PDF
23. Rapid identification of genomic structural variations with nanopore sequencing enables blood-based cancer monitoring
- Author
-
Martijn P. Lolkema, Maurice P.H.M. Jansen, Christina Stangl, Wigard P. Kloosterman, Jean C. A. Helmijr, Ivo Renkens, Markus J. van Roosmalen, Lisanne F. van Dessel, Anouk C. de Jong, John W.M. Martens, Sam de Blank, Jose Espejo Valle-Inclan, and Chris J. de Witte
- Subjects
Rapid identification ,Prostate cancer ,Somatic cell ,Breakpoint ,medicine ,Cancer ,Biomarker (medicine) ,Digital polymerase chain reaction ,Nanopore sequencing ,Computational biology ,Biology ,medicine.disease - Abstract
Somatic genomic structural variations (SVs) are promising personalized biomarkers for sensitive and specific detection of circulating tumor DNA (ctDNA) in liquid biopsies. However, affordable and fast identification of such SV biomarkers is challenging, which hinders routine use in the clinic. Here, we developed a novel approach - termed SHARC - for rapid discovery of somatic SVs as personalized tumor biomarkers. SHARC combines low coverage cancer genome sketching by using Oxford Nanopore sequencing with random forest classification and a dedicated filtering pipeline to enrich for somatic SVs. Our method leverages the real-time and long-read capabilities of nanopore sequencing to identify somatic SV breakpoints at nucleotide resolution from a tumor tissue biopsy within three days. We applied SHARC to tumor samples of high-grade ovarian and prostate cancer and validated on average ten somatic SVs per sample with PCR mini-amplicons. An accompanying method for SV breakpoint detection from liquid biopsies was devised based on digital PCR, enabling detection of cancer in a quantitative manner. Using this method, we retrospectively monitored treatment response in patients with metastatic prostate cancer. Our work demonstrates that SHARC forms a universal framework for rapid development of personalized biomarker assays for blood-based monitoring of any cancer type.
- Published
- 2019
- Full Text
- View/download PDF
24. P174 Generation of organoids from ovarian adult granulosa cell tumours for individualized drug screening
- Author
-
Ronald P. Zweemer, Ferdinando Sereno, Petronella O. Witteveen, Joline F. Roze, Christina Stangl, J. W. Groeneweg, H. S. van Meurs, S. T. Paijens, Hans W. Nijman, Jmj Piek, Geertruida N. Jonges, L Van Lonkhuijzen, Glen R. Monroe, Christianne A.R. Lok, and Ellen Stelloo
- Subjects
Chemotherapy ,business.industry ,medicine.medical_treatment ,Granulosa cell ,medicine.disease ,Carboplatin ,chemistry.chemical_compound ,Paclitaxel ,chemistry ,Cell culture ,Cancer research ,Organoid ,Medicine ,business ,Ovarian cancer ,Tamoxifen ,medicine.drug - Abstract
Introduction/Background Adult granulosa cell tumours (aGCT) constitute a rare subtype of ovarian cancer, with >90% of tumours characterized by the FOXL2 c.402C>G mutation. Recurrences occur in nearly 50% of patients and are associated with a poor prognosis. Surgery is the mainstay of treatment, since the effect of adjuvant therapies are limited. Chemotherapy and hormone therapy response is difficult to predict and patient numbers insufficient to conduct informative clinical trials. Patient-specific aGCT organoid and 2D culture establishment could evaluate the effect of novel therapeutic options on a relatively large scale and enable personalized treatment in this neglected patient group. Methodology Samples from 46 tumours (4 primary and 42 recurrences) from 18 patients were collected, mechanically homogenized to single cells and small tissue pieces, and seeded in 50% Cultrex Basement Membrane Extract on day of surgery. Basal medium (DMEM/F12) supplemented with various growth factor combinations promoted organoid formation. Medium was refreshed every 3–4 days and cells passaged every 3 weeks. In parallel, 2D monolayer cell cultures were established to perform drug screens for later validation in the more physiologically relevant organoids as they became available. Organoid and 2D tumour origin verification is ongoing by FOXL2 c.402C>G PCR. Results Tumour tissue cultivation resulted in 3D structures that remained viable for 3–4 passages, with an establishment rate of approximately 75% in primary and 26% in recurrences. Morphology of masses ranged from grape-like to cystic, with primary organoids demonstrating a more cystic morphology (figure 1) that could be robustly passaged. Monolayer cultures in DMEM/F12/FBS were successfully established for future drug screening with carboplatin, paclitaxel, tamoxifen and other first-line treatments. Conclusion Patient-derived aGCT organoids can be established. Organoid establishment is more successful for primary tumours than for metastases, and organoid establishment optimization is necessary. Tumour cell culture establishment and drug screens are likely to provide insight into patient-specific treatment response. Disclosure Nothing to disclose.
- Published
- 2019
- Full Text
- View/download PDF
25. Partner-independent fusion gene detection by multiplexed CRISPR/Cas9 enrichment and long-read Nanopore sequencing
- Author
-
Tamara Verbeek, Gijs van Haaften, Ivo Renkens, Markus J. van Roosmalen, Emile E. Voest, Jose Espejo Valle-Inclan, Wigard P. Kloosterman, Christina Stangl, Anton G. Henssen, Sam de Blank, Ronald W. Stam, Rocío Chamorro González, and Glen R. Monroe
- Subjects
Fusion gene ,KMT2A ,Cas9 ,Breakpoint ,biology.protein ,CRISPR ,Nanopore sequencing ,Computational biology ,Biology ,Gene ,Genome - Abstract
Fusion genes are hallmarks of various cancer types and important determinants for diagnosis, prognosis and treatment possibilities. The promiscuity of fusion genes with respect to partner choice and exact breakpoint-positions restricts their detection in the diagnostic setting, even for known and recurrent fusion gene configurations. To accurately identify these gene fusions in an unbiased manner, we developed FUDGE: a FUsion gene Detection assay from Gene Enrichment. FUDGE couples target-selected and strand-specific CRISPR/Cas9 activity for enrichment and detection of fusion gene drivers (e.g. BRAF, EWSR1, KMT2A/MLL) - without prior knowledge of fusion partner or breakpoint-location - to long-read Nanopore sequencing. FUDGE encompasses a dedicated bioinformatics approach (NanoFG) to detect fusion genes from Nanopore sequencing data. Our strategy is flexible with respect to target choice and enables multiplexed enrichment for simultaneous analysis of several genes in multiple samples in a single sequencing run. We observe on average a 508 fold on-target enrichment and identify fusion breakpoints at nucleotide resolution - all within two days. We demonstrate that FUDGE effectively identifies fusion genes in cancer cell lines, tumor samples and on whole genome amplified DNA irrespective of partner gene or breakpoint-position in 100% of cases. Furthermore, we show that FUDGE is superior to routine diagnostic methods for fusion gene detection. In summary, we have developed a rapid and versatile fusion gene detection assay, providing an unparalleled opportunity for pan-cancer detection of fusion genes in routine diagnostics.
- Published
- 2019
- Full Text
- View/download PDF
26. Diverse
- Author
-
Christina, Stangl, Jasmin B, Post, Markus J, van Roosmalen, Nizar, Hami, Ingrid, Verlaan-Klink, Harmjan R, Vos, Robert M, van Es, Marco J, Koudijs, Emile E, Voest, Hugo J G, Snippert, and W P, Kloosterman
- Subjects
ErbB Receptors ,Organoids ,Proto-Oncogene Proteins B-raf ,HEK293 Cells ,Drug Resistance, Neoplasm ,MAP Kinase Signaling System ,Humans ,Cell Differentiation ,Oncogene Fusion ,Molecular Targeted Therapy ,Colorectal Neoplasms ,MAP Kinase Kinase Kinases ,Protein Kinase Inhibitors - Abstract
Fusion genes can be oncogenic drivers in a variety of cancer types and represent potential targets for targeted therapy. The
- Published
- 2019
27. Type-7 metabotropic glutamate receptors negatively regulate α
- Author
-
Luisa, Iacovelli, Luisa, Di Menna, Daniel, Peterlik, Christina, Stangl, Rosamaria, Orlando, Gemma, Molinaro, Antonio, De Blasi, Valeria, Bruno, Giuseppe, Battaglia, Peter J, Flor, Nicole, Uschold-Schmidt, and Ferdinando, Nicoletti
- Subjects
Male ,Mice, Knockout ,Mice, 129 Strain ,Dose-Response Relationship, Drug ,Receptors, Metabotropic Glutamate ,Mice, Inbred C57BL ,Mice ,HEK293 Cells ,Receptors, Adrenergic, alpha-1 ,Excitatory Amino Acid Agonists ,Animals ,Humans ,Adrenergic alpha-1 Receptor Agonists ,Signal Transduction - Abstract
We studied the interaction between mGlu7 and α
- Published
- 2016
28. Blocking metabotropic glutamate receptor subtype 5 relieves maladaptive chronic stress consequences
- Author
-
Peter J. Flor, Dominic Schmidt, Amelie Bauer, Stefan O. Reber, Franziska Zajicek, Nicole Uschold-Schmidt, Daniel Peterlik, Anna Bludau, Dominik Grabski, Jana Keller, Christina Stangl, Tobias Killian, Georg Jaeschke, and Lothar Lindemann
- Subjects
0301 basic medicine ,Dominance-Subordination ,Male ,Allosteric modulator ,Fever ,Hydrocortisone ,Pyridines ,Receptor, Metabotropic Glutamate 5 ,Immunology ,Context (language use) ,Anxiety ,Bioinformatics ,Social Environment ,03 medical and health sciences ,Behavioral Neuroscience ,Mice ,0302 clinical medicine ,Adrenocorticotropic Hormone ,medicine ,Animals ,Chronic stress ,Mice, Knockout ,Dose-Response Relationship, Drug ,Endocrine and Autonomic Systems ,Imidazoles ,medicine.disease ,Up-Regulation ,Mice, Inbred C57BL ,030104 developmental biology ,Prenatal stress ,Metabotropic glutamate receptor ,Chronic Disease ,Major depressive disorder ,medicine.symptom ,Psychology ,Psychosocial ,030217 neurology & neurosurgery ,Stress, Psychological ,Clinical psychology - Abstract
Etiology and pharmacotherapy of stress-related psychiatric conditions and somatoform disorders are areas of high unmet medical need. Stressors holding chronic plus psychosocial components thereby bear the highest health risk. Although the metabotropic glutamate receptor subtype 5 (mGlu5) is well studied in the context of acute stress-induced behaviors and physiology, virtually nothing is known about its potential involvement in chronic psychosocial stress. Using the mGlu5 negative allosteric modulator CTEP (2-chloro-4-[2-[2,5-dimethyl-1-[4-(trifluoromethoxy)phenyl]imidazol-4yl]ethynyl]pyridine), a close analogue of the clinically active drug basimglurant – but optimized for rodent studies, as well as mGlu5-deficient mice in combination with a mouse model of male subordination (termed CSC, chronic subordinate colony housing), we demonstrate that mGlu5 mediates multiple physiological, immunological, and behavioral consequences of chronic psychosocial stressor exposure. For instance, CTEP dose-dependently relieved hypothalamo-pituitary-adrenal axis dysfunctions, colonic inflammation as well as the CSC-induced increase in innate anxiety; genetic ablation of mGlu5 in mice largely reproduced the stress-protective effects of CTEP and additionally ameliorated CSC-induced physiological anxiety. Interestingly, CSC also induced an upregulation of mGlu5 in the hippocampus, a stress-regulating brain area. Taken together, our findings provide evidence that mGlu5 is an important mediator for a wide range of chronic psychosocial stress-induced alterations and a potentially valuable drug target for the treatment of chronic stress-related pathologies in man.
- Published
- 2016
29. Abstract 490: Identification of oncogenic gene fusions in primary colon cancers
- Author
-
Katharina Biermann, Markus J. van Roosmalen, Zarina S. Lalmahomed, Christina Stangl, Anieta M. Sieuwerts, Marco J. Koudijs, John W.M. Martens, Anne van Galen, Fried J. T. Zwartkruis, John A. Foekens, Armel Lefebvre, Jan N. M. IJzermans, Salo N. Ooft, Marcel Smid, Ronne Brunekreef, Wigard P. Kloosterman, Emile E. Voest, Robert R. J. Coebergh van den Braak, and Mark Pieterse
- Subjects
Cancer Research ,Primary (chemistry) ,Oncology ,Cancer research ,Identification (biology) ,Biology ,Gene - Abstract
Introduction: Fusion genes (such as ALK-EML) are potentially attractive targets for anti-cancer treatment. In the era of personalized medicine exploring the identification, incidence, and functionality of fusion genes may contribute to effective treatment approaches. Methods: We performed a comprehensive and unbiased screening for gene fusions in a clinically well-defined prospectively collected cohort of 278 primary stage I to III colon cancers. Illumina RNA sequencing was performed using RNA from the fresh frozen samples. The STAR fusion gene detection pipeline and GATK RNA-seq variant calling were used to identify fusion genes and detect somatic genetic variations. Gene fusions were considered relevant when recurrent, when resulting in divergent expression (outlier analysis) or when functional relevance was predicted (i.e. kinase fusions). Results: 2.5% of all samples contained a relevant gene fusion. Kinase fusions were most prevalent with a frequency of 1.8%. Three BRAF-fusions were identified, two known (TRIM24-BRAF & AGAP3-BRAF) and one novel fusion (DLG1-BRAF). Co-expression with ERK1 in HEK293 cells resulted in enhanced EKR1 phosphorylation. The outlier analysis revealed four unique fusions (EML4-NTRK3, RRBP1-RET, USPX9-ERAS & EIF3E-RSPO2). The EML4-NTRK3 fusion led to increased expression of the tyrosine kinase encoding domain exons, which is retained in the fusion transcript. Co-expression with ERK1 in HEK293 cells resulted in enhanced EKR1 phosphorylation. The tumor with the RRBP1-RET fusion was pan negative for known driver mutations, as reported in literature. Mate-pair sequencing of the USPX9-ERAS fusion gene region revealed that the fusion gene was caused at the genomic level by a highly local chromothripsis event on chromosome X spanning solely the region covered by USP9X and ERAS, leading to high ERAS expression. ERAS is a constitutionally active RAS protein and normally only expressed in embryonic stem cells. Analysis of phosphorylated AKT after expression of the USP9X-ERAS fusion gene in NIH-3T3 A14 cells showed activation of AKT signaling. The occurrence of one R-spondin fusion in our set (0.4%) is low compared to the previously described 9.5%, which may be explained by difference in incidence of KRAS- or BRAF-mutations in the two cohorts, since all R-spondin fusions occurred in a tumor with a KRAS- or BRAF-mutation. All fusion genes were expressed in cell lines resulting in the activation of AKT signaling. Finally, oncogenic fusions were only found in lymph node negative tumors and were not observed in tumors with classical driver mutations in BRAF and KRAS except for the R-spondin fusion. Conclusions: We found oncogenic gene fusions including novel fusions with a frequency of 2.5%, including an USP9X-ERAS fusion with strong oncogenic activity in vitro. Although recurrent fusion genes are rare events in colorectal cancer they may represent functional drivers and provide potential novel leads for personalized therapeutic strategies. Citation Format: Robert R. Coebergh van den Braak, Wigard P. Kloosterman, Mark Pieterse, Markus van Roosmalen, Anieta M. Sieuwerts, Christina Stangl, Ronne Brunekreef, Zarina S. Lalmahomed, Salo Ooft, Anne van Galen, Marcel Smid, Armel Lefebvre, Fried J. Zwartkruis, John W. Martens, John A. Foekens, Katharina Biermann, Marco J. Koudijs, Jan N. IJzermans, Emile E. Voest. Identification of oncogenic gene fusions in primary colon cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 490. doi:10.1158/1538-7445.AM2017-490
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.