37 results on '"Azoitei, Ninel"'
Search Results
2. Protein kinase D2: a versatile player in cancer biology
- Author
-
Azoitei, Ninel, Cobbaut, Mathias, Becher, Alexander, Van Lint, Johan, and Seufferlein, Thomas
- Published
- 2018
- Full Text
- View/download PDF
3. Physical plasma-triggered ROS induces tumor cell death upon cleavage of HSP90 chaperone
- Author
-
Bekeschus, Sander, Lippert, Maxi, Diepold, Kristina, Chiosis, Gabriela, Seufferlein, Thomas, and Azoitei, Ninel
- Published
- 2019
- Full Text
- View/download PDF
4. DNA methylation‐associated allelic inactivation regulates Keratin 19 gene expression during pancreatic development and carcinogenesis.
- Author
-
Krüger, Jana, Fischer, Anja, Breunig, Markus, Allgöwer, Chantal, Schulte, Lucas, Merkle, Jessica, Mulaw, Medhanie A, Okeke, Nnamdi, Melzer, Michael K, Morgenstern, Clara, Azoitei, Ninel, Seufferlein, Thomas, Barth, Thomas FE, Siebert, Reiner, Hohwieler, Meike, and Kleger, Alexander
- Subjects
GENE expression ,PLURIPOTENT stem cells ,DNA methyltransferases ,HUMAN stem cells ,PANCREATIC duct ,KERATIN ,DNA methylation ,DEMETHYLATION ,PANCREATIC enzymes - Abstract
Within the pancreas, Keratin 19 (KRT19) labels the ductal lineage and is a determinant of pancreatic ductal adenocarcinoma (PDAC). To investigate KRT19 expression dynamics, we developed a human pluripotent stem cell (PSC)‐based KRT19‐mCherry reporter system in different genetic backgrounds to monitor KRT19 expression from its endogenous gene locus. A differentiation protocol to generate mature pancreatic duct‐like organoids was applied. While KRT19/mCherry expression became evident at the early endoderm stage, mCherry signal was present in nearly all cells at the pancreatic endoderm (PE) and pancreatic progenitor (PP) stages. Interestingly, despite homogenous KRT19 expression, mCherry positivity dropped to 50% after ductal maturation, indicating a permanent switch from biallelic to monoallelic expression. DNA methylation profiling separated the distinct differentiation intermediates, with site‐specific DNA methylation patterns occurring at the KRT19 locus during ductal maturation. Accordingly, the monoallelic switch was partially reverted upon treatment with a DNA‐methyltransferase inhibitor. In human PDAC cohorts, high KRT19 levels correlate with low locus methylation and decreased survival. At the same time, activation of oncogenic KRASG12D signalling in our reporter system reversed monoallelic back to biallelic KRT19 expression in pancreatic duct‐like organoids. Allelic reactivation was also detected in single‐cell transcriptomes of human PDACs, which further revealed a positive correlation between KRT19 and KRAS expression. Accordingly, KRAS mutant PDACs had higher KRT19 mRNA but lower KRT19 gene locus DNA methylation than wildtype counterparts. KRT19 protein was additionally detected in plasma of PDAC patients, with higher concentrations correlating with shorter progression‐free survival in gemcitabine/nabPaclitaxel‐treated and opposing trends in FOLFIRINOX‐treated patients. Apart from being an important pancreatic ductal lineage marker, KRT19 appears tightly controlled via a switch from biallelic to monoallelic expression during ductal lineage entry and is aberrantly expressed after oncogenic KRASG12D expression, indicating a role in PDAC development and malignancy. Soluble KRT19 might serve as a relevant biomarker to stratify treatment. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. BTK Isoforms p80 and p65 Are Expressed in Head and Neck Squamous Cell Carcinoma (HNSCC) and Involved in Tumor Progression.
- Author
-
Betzler, Annika C., Strobel, Hannah, Abou Kors, Tsima, Ezić, Jasmin, Lesakova, Kristina, Pscheid, Ronja, Azoitei, Ninel, Sporleder, Johanna, Staufenberg, Anna-Rebekka, Drees, Robert, Weissinger, Stephanie E., Greve, Jens, Doescher, Johannes, Theodoraki, Marie-Nicole, Schuler, Patrick J., Laban, Simon, Kibe, Toshiro, Kishida, Michiko, Kishida, Shosei, and Idel, Christian
- Subjects
DISEASE progression ,CARCINOGENESIS ,NUCLEAR proteins ,HEAD & neck cancer ,GENE expression ,PROTEIN-tyrosine kinases ,MESSENGER RNA ,METHYLATION ,CELL proliferation ,SQUAMOUS cell carcinoma ,EPIGENOMICS - Abstract
Simple Summary: Bruton's Tyrosine Kinase (BTK) was originally considered to be primarily expressed in cells of hematopoietic origin. Apart from the 77 kDa BTK isoform expressed in immune cells, elevated expression of novel BTK isoforms of 80 and 65 kDa have been recently described for several solid tumor entities. These newly described isoforms have been linked to tumor growth and poor prognosis. Therefore, we aimed to investigate whether BTK isoforms are also expressed in head and neck squamous cell carcinoma (HNSCC) and further the molecular and cellular consequences of BTK expression for HNSCC tumorigenesis. We confirmed the expression of the BTK-p65 and BTK-p80 isoforms in HNSCC and revealed that both isoforms are products of the same mRNA. Abrogation of BTK activity inhibited tumor progression in our study. Thus, targeting BTK activity appears as a promising therapeutic option for patients suffering from BTK expressing HNSCC. Here, we describe the expression of Bruton's Tyrosine Kinase (BTK) in head and neck squamous cell carcinoma (HNSCC) cell lines as well as in primary HNSCC samples. BTK is a kinase initially thought to be expressed exclusively in cells of hematopoietic origin. Apart from the 77 kDa BTK isoform expressed in immune cells, particularly in B cells, we identified the 80 kDa and 65 kDa BTK isoforms in HNSCC, recently described as oncogenic. Importantly, we revealed that both isoforms are products of the same mRNA. By investigating the mechanism regulating oncogenic BTK-p80/p65 expression in HNSSC versus healthy or benign tissues, our data suggests that the epigenetic process of methylation might be responsible for the initiation of BTK-p80/p65 expression in HNSCC. Our findings demonstrate that chemical or genetic abrogation of BTK activity leads to inhibition of tumor progression in terms of proliferation and vascularization in vitro and in vivo. These observations were associated with cell cycle arrest and increased apoptosis and autophagy. Together, these data indicate BTK-p80 and BTK-p65 as novel HNSCC-associated oncogenes. Owing to the fact that abundant BTK expression is a characteristic feature of primary and metastatic HNSCC, targeting BTK activity appears as a promising therapeutic option for HNSCC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Protein kinase D2 is a crucial regulator of tumour cell-endothelial cell communication in gastrointestinal tumours
- Author
-
Azoitei, Ninel, Pusapati, Ganesh Varma, Kleger, Alexander, Moller, Peter, Kufer, Rainer, Genze, Felicitas, Wagner, Martin, van Lint, Johan, Carmeliet, Peter, Adler, Guido, and Seufferlein, Thomas
- Subjects
Protein kinases -- Research ,Protein kinases -- Physiological aspects ,Gastrointestinal tumors -- Diagnosis ,Gastrointestinal tumors -- Genetic aspects ,Cell proliferation -- Research ,Cell proliferation -- Physiological aspects ,Neovascularization -- Research ,Neovascularization -- Physiological aspects ,Health - Published
- 2010
7. Organoids at the PUB: The Porcine Urinary Bladder Serves as a Pancreatic Niche for Advanced Cancer Modeling.
- Author
-
Melzer, Michael Karl, Breunig, Markus, Arnold, Frank, Wezel, Felix, Azoitei, Anca, Roger, Elodie, Krüger, Jana, Merkle, Jessica, Schütte, Lena, Resheq, Yazid, Hänle, Mark, Zehe, Viktor, Zengerling, Friedemann, Azoitei, Ninel, Klein, Lukas, Penz, Frederike, Singh, Shiv K., Seufferlein, Thomas, Hohwieler, Meike, and Bolenz, Christian
- Published
- 2022
- Full Text
- View/download PDF
8. Activation of the IκB kinase complex is sufficient for neuronal differentiation of PC12 cells
- Author
-
Azoitei, Ninel, Wirth, Thomas, and Baumann, Bernd
- Published
- 2005
9. NF-κB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression
- Author
-
Huber, Margit A., Azoitei, Ninel, Baumann, Bernd, Grünert, Stefan, Sommer, Andreas, Pehamberger, Hubert, Kraut, Norbert, Beug, Hartmut, and Wirth, Thomas
- Published
- 2004
10. Functional Genomic Screening During Somatic Cell Reprogramming Identifies DKK3 as a Roadblock of Organ Regeneration.
- Author
-
Arnold, Frank, Mahaddalkar, Pallavi U, Kraus, Johann M., Zhong, Xiaowei, Bergmann, Wendy, Srinivasan, Dharini, Gout, Johann, Roger, Elodie, Beutel, Alica K., Zizer, Eugen, Tharehalli, Umesh, Daiss, Nora, Russell, Ronan, Perkhofer, Lukas, Oellinger, Rupert, Lin, Qiong, Azoitei, Ninel, Weiss, Frank‐Ulrich, Lerch, Markus M., and Liebau, Stefan
- Subjects
PLURIPOTENT stem cells ,SOMATIC cells ,REGENERATION (Biology) ,INDUCED pluripotent stem cells ,EMBRYONIC stem cells ,LIVER cells - Abstract
Somatic cell reprogramming and tissue repair share relevant factors and molecular programs. Here, Dickkopf‐3 (DKK3) is identified as novel factor for organ regeneration using combined transcription‐factor‐induced reprogramming and RNA‐interference techniques. Loss of Dkk3 enhances the generation of induced pluripotent stem cells but does not affect de novo derivation of embryonic stem cells, three‐germ‐layer differentiation or colony formation capacity of liver and pancreatic organoids. However, DKK3 expression levels in wildtype animals and serum levels in human patients are elevated upon injury. Accordingly, Dkk3‐null mice display less liver damage upon acute and chronic failure mediated by increased proliferation in hepatocytes and LGR5+ liver progenitor cell population, respectively. Similarly, recovery from experimental pancreatitis is accelerated. Regeneration onset occurs in the acinar compartment accompanied by virtually abolished canonical‐Wnt‐signaling in Dkk3‐null animals. This results in reduced expression of the Hedgehog repressor Gli3 and increased Hedgehog‐signaling activity upon Dkk3 loss. Collectively, these data reveal Dkk3 as a key regulator of organ regeneration via a direct, previously unacknowledged link between DKK3, canonical‐Wnt‐, and Hedgehog‐signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. Human Pluripotent Stem Cells Go Diabetic: A Glimpse on Monogenic Variants.
- Author
-
Heller, Sandra, Melzer, Michael Karl, Azoitei, Ninel, Julier, Cécile, and Kleger, Alexander
- Subjects
PLURIPOTENT stem cells ,HUMAN stem cells ,OCCUPATIONAL diseases ,TYPE 1 diabetes ,DIABETES ,TYPE 2 diabetes - Abstract
Diabetes, as one of the major diseases in industrial countries, affects over 350 million people worldwide. Type 1 (T1D) and type 2 diabetes (T2D) are the most common forms with both types having invariable genetic influence. It is accepted that a subset of all diabetes patients, generally estimated to account for 1–2% of all diabetic cases, is attributed to mutations in single genes. As only a subset of these genes has been identified and fully characterized, there is a dramatic need to understand the pathophysiological impact of genetic determinants on β -cell function and pancreatic development but also on cell replacement therapies. Pluripotent stem cells differentiated along the pancreatic lineage provide a valuable research platform to study such genes. This review summarizes current perspectives in applying this platform to study monogenic diabetes variants. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. Allelic regulation of Keratin 19 gene expression during pancreatic development and carcinogenesis.
- Author
-
Krüger, Jana, Fischer, Anja, Breunig, Markus, Allgöwer, Chantal, Schulte, Lucas, Merkle, Jessica, Mulaw, Medhanie, Okeke, Nnamdi, Melzer, Michael K., Azoitei, Ninel, Seufferlein, Thomas, Siebert, Reiner, Hohwieler, Meike, and Kleger, Alexander
- Published
- 2023
- Full Text
- View/download PDF
13. Enteropathogenic Infections: Organoids Go Bacterial.
- Author
-
Hentschel, Viktoria, Arnold, Frank, Seufferlein, Thomas, Azoitei, Ninel, Kleger, Alexander, and Müller, Martin
- Subjects
ORGANOIDS ,HYGIENE ,INTESTINAL mucosa ,CELL anatomy ,MEDICAL care ,INTESTINAL infections ,TISSUE engineering - Abstract
Enteric infections represent a major health care challenge which is particularly prevalent in countries with restricted access to clean water and sanitation and lacking personal hygiene precautions, altogether facilitating fecal-oral transmission of a heterogeneous spectrum of enteropathogenic microorganisms. Among these, bacterial species are responsible for a considerable proportion of illnesses, hospitalizations, and fatal cases, all of which have been continuously contributing to ignite researchers' interest in further exploring their individual pathogenicity. Beyond the universally accepted animal models, intestinal organoids are increasingly valued for their ability to mimic key architectural and physiologic features of the native intestinal mucosa. As a consequence, they are regarded as the most versatile and naturalistic in vitro model of the gut, allowing monitoring of adherence, invasion, intracellular trafficking, and propagation as well as repurposing components of the host cell equipment. At the same time, infected intestinal organoids allow close characterization of the host epithelium's immune response to enteropathogens. In this review, (i) we provide a profound update on intestinal organoid-based tissue engineering, (ii) we report the latest pathophysiological findings defining the infected intestinal organoids, and (iii) we discuss the advantages and limitations of this in vitro model. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
14. Sildenafil triggers tumor lethality through altered expression of HSP90 and degradation of PKD2.
- Author
-
Chen, Lu, Liu, Yang, Becher, Alexander, Diepold, Kristina, Schmid, Evi, Fehn, Adrian, Brunner, Cornelia, Rouhi, Arefeh, Chiosis, Gabriela, Cronauer, Marcus, Seufferlein, Thomas, and Azoitei, Ninel
- Subjects
SILDENAFIL ,CANCER cell proliferation ,PHOSPHODIESTERASE-5 inhibitors ,PROTEOLYSIS ,PROTEIN kinases - Abstract
The repurposing of existing drugs has emerged as an attractive additional strategy to the development of novel compounds in the fight against cancerous diseases. Inhibition of phosphodiesterase 5 (PDE5) has been claimed as a potential approach to target various cancer subtypes in recent years. However, data on the treatment of tumors with PDE5 inhibitors as well as the underlying mechanisms are as yet very scarce. Here, we report that treatment of tumor cells with low concentrations of Sildenafil was associated with decreased cancer cell proliferation and augmented apoptosis in vitro and resulted in impaired tumor growth in vivo. Notably, incubation of cancer cells with Sildenafil was associated with altered expression of HSP90 chaperone followed by degradation of protein kinase D2, a client protein previously reported to be involved in tumor growth. Furthermore, the involvement of low doses of PU-H71, an HSP90 inhibitor currently under clinical evaluation, in combination with low concentrations of Sildenafil, synergistically and negatively impacted on the viability of cancer cells in vivo. Taken together, our study suggests that repurposing of already approved drugs, alone or in combination with oncology-dedicated compounds, may represent a novel cancer therapeutic strategy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
15. Adenosine receptor 2B activity promotes autonomous growth, migration as well as vascularization of head and neck squamous cell carcinoma cells.
- Author
-
Wilkat, Max, Bast, Hanna, Drees, Robert, Dünser, Johannes, Mahr, Amelie, Azoitei, Ninel, Marienfeld, Ralf, Frank, Felicia, Brhel, Magnus, Ushmorov, Alexey, Greve, Jens, Goldberg‐Bockhorn, Eva, Theodoraki, Marie‐Nicole, Doescher, Johannes, Laban, Simon, Schuler, Patrick J., Hoffmann, Thomas K., and Brunner, Cornelia
- Subjects
SQUAMOUS cell carcinoma ,ADENOSINES ,INHIBITION of cellular proliferation ,CELL receptors ,CELL physiology - Abstract
Adenosine is a signaling molecule that exerts dual effects on tumor growth: while it inhibits immune cell function and thereby prevents surveillance by the immune system, it influences tumorigenesis directly via activation of adenosine receptors on tumor cells at the same time. However, the adenosine‐mediated mechanisms affecting oncogenic processes particularly in head and neck squamous cell carcinomas (HNSCC) are not fully understood. Here, we investigated the role of adenosine receptor activity on HNSCC‐derived cell lines. Targeting the adenosine receptor A2B (ADORA2B) on these cells with the inverse agonist/antagonist PSB‐603 leads to inhibition of cell proliferation, transmigration as well as VEGFA secretion in vitro. At the molecular level, these effects were associated with cell cycle arrest as well as the induction of the apoptotic pathway. In addition, shRNA‐mediated downmodulation of ADORA2B expression caused decreased proliferation. Moreover, in in vivo xenograft experiments, chemical and genetic abrogation of ADORA2B activity impaired tumor growth associated with decreased tumor vascularization. Together, our findings characterize ADORA2B as a crucial player in the maintenance of HNSCC and, therefore, as a potential therapeutic target for HNSCC treatment. What's new? The adenosinergic system plays an important role in the development of many types of cancer. However, the underlying mechanisms remain poorly understood. This study set to identify the oncogenic function of the adenosine receptor 2B (ADORA2B) in head and neck squamous cell carcinoma (HNSCC)‐derived tumor cells. The results show that ADORA2B is upregulated and constitutively active in HNSCC‐derived cell lines. This activity is sufficient to promote autonomous cell growth, migration, and angiogenesis in vitro and in vivo. The data suggest ADORA2B as an important biomarker and potential therapeutic target for the treatment of HNSCC and other ADORA2B‐expressing solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
16. Die BTK-Isoformen p80 und p65 werden im Kopf-Hals-Plattenepithelkarzinom (HNSCC) exprimiert und sind an der Tumorprogression beteiligt.
- Author
-
Betzler, AnnikaC, Strobel, Hannah, Abou, Tsima Kors, Azoitei, Ninel, Greve, Jens, Theodoraki, Marie-Nicole, Schuler, PatrickJ, Laban, Simon, Hoffmann, ThomasK, Grassilli, Emanuela, and Brunner, Cornelia
- Published
- 2023
- Full Text
- View/download PDF
17. Requirement for YAP1 signaling in myxoid liposarcoma.
- Author
-
Trautmann, Marcel, Cheng, Ya‐Yun, Jensen, Patrizia, Azoitei, Ninel, Brunner, Ines, Hüllein, Jennifer, Slabicki, Mikolaj, Isfort, Ilka, Cyra, Magdalene, Berthold, Ruth, Wardelmann, Eva, Huss, Sebastian, Altvater, Bianca, Rossig, Claudia, Hafner, Susanne, Simmet, Thomas, Ståhlberg, Anders, Åman, Pierre, Zenz, Thorsten, and Lange, Undine
- Abstract
Myxoid liposarcomas (MLS), malignant tumors of adipocyte origin, are driven by the FUS‐DDIT3 fusion gene encoding an aberrant transcription factor. The mechanisms whereby FUS‐DDIT3 mediates sarcomagenesis are incompletely understood, and strategies to selectively target MLS cells remain elusive. Here we show, using an unbiased functional genomic approach, that FUS‐DDIT3‐expressing mesenchymal stem cells and MLS cell lines are dependent on YAP1, a transcriptional co‐activator and central effector of the Hippo pathway involved in tissue growth and tumorigenesis, and that increased YAP1 activity is a hallmark of human MLS. Mechanistically, FUS‐DDIT3 promotes YAP1 expression, nuclear localization, and transcriptional activity and physically associates with YAP1 in the nucleus of MLS cells. Pharmacologic inhibition of YAP1 activity impairs the growth of MLS cells in vitro and in vivo. These findings identify overactive YAP1 signaling as unifying feature of MLS development that could represent a novel target for therapeutic intervention. Synopsis: The transcriptional co‐activator YAP1 is essential in myxoid liposarcoma (MLS), an aggressive soft‐tissue tumor. This study reveals a link between aberrant YAP1 signaling and the FUS DDIT3 fusion oncoprotein that drives MLS. Pharmacologic YAP1 inhibition impairs MLS growth in vitro and in vivo. YAP1, encoding a transcriptional co activator that is inhibited by the Hippo pathway, was identified by RNA interference screening as an essential gene in mesenchymal stem cells expressing FUS‐DDIT3.FUS‐DDIT3‐expressing MLS cell lines and MLS patient specimens exhibited increased YAP1 activity, and YAP1 suppression in MLS cells caused proliferation arrest, senescence, and apoptosis.FUS DDIT3 induced the expression and nuclear localization of YAP1 and its downstream effectors.FUS‐DDIT3 and YAP1 physically associated in the nucleus of MLS cells.MLS cells were sensitive to pharmacologic blockade of YAP1 activity. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
18. BTK isoforms p80 and p65 are expressed in head and neck squamous cell carcinoma (HNSCC) and involved in tumor progression.
- Author
-
Betzler, AnnikaC, Strobel, Hannah, Abou, Tsima Kors, Azoitei, Ninel, Greve, Jens, Theodoraki, Marie-Nicole, Schuler, PatrickJ, Laban, Simon, Hoffmann, ThomasK, Grassilli, Emanuela, and Brunner, Cornelia
- Published
- 2023
- Full Text
- View/download PDF
19. PKM2 promotes tumor angiogenesis by regulating HIF-1α through NF-κB activation.
- Author
-
Azoitei, Ninel, Becher, Alexander, Steinestel, Konrad, Rouhi, Arefeh, Diepold, Kristina, Genze, Felicitas, Simmet, Thomas, and Seufferlein, Thomas
- Subjects
- *
PYRUVATE kinase , *NEOVASCULARIZATION inhibitors , *GLYCOLYSIS , *CANCER cells , *CELL cycle , *CELL proliferation , *APOPTOSIS - Abstract
Background: Initially identified as a molecule that regulates the final step of glycolysis, the M2 isoform of pyruvate kinase (PKM2) was recently reported to have a central role in the metabolic reprogramming of cancer cells as well as participating in cell cycle progression and gene transcription. Despite intensive efforts, the intricate molecular mechanisms through which PKM2 regulates tumor progression remain elusive. Methods: The proliferation and apoptosis of various pancreatic cancer cells using lentiviral-mediated PKM2 abrogation were assessed in vitro via Western blot and flow cytometric assay while the in vivo experiments involved tumor xenograft on chicken chorionallantoic membranes and immunohistochemistry on human tissue specimens. In order to decipher the molecular mechanism of HIF-1α and p65/RelA regulation by PKM2 in cancer cells cultivated in hypoxic atmosphere or normoxia we involved various biochemical assays such as Western blotting, immunoprecipitation, reporter gene assay and ELISA. Results: Strong expression of PKM2 was observed in 68 % of human pancreatic adenocarcinoma specimens and almost all analyzed pancreatic cancer cell lines. Abrogation of PKM2 resulted in impaired proliferation and augmented apoptosis in vitro as well as impaired tumor growth and decreased blood vessel formation in vivo. Furthermore, deletion of PKM2 negatively impacted hypoxia-induced HIF-1α accumulation and promoter activity ultimately resulting in impaired secretion of VEGF. Conclusions: Our study suggests that in hypoxic pancreatic tumors PKM2 interferes both with NF-κB/p65 and HIF-1α activation that ultimately triggers VEGF-A secretion and subsequent blood vessel formation. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
20. HSP90 Supports Tumor Growth and Angiogenesis through PRKD2 Protein Stabilization.
- Author
-
Azoitei, Ninel, Diepold, Kristina, Brunner, Cornelia, Rouhi, Arefeh, Genze, Felicitas, Becher, Alexander, Kestler, Hans, van Lint, Johan, Chiosis, Gabriela, Koren III, John, Fröhling, Stefan, Scholl, Claudia, and Seufferlein, Thomas
- Subjects
- *
TUMOR growth , *NEOVASCULARIZATION , *PROTEIN stability , *CANCER cells , *GASTROINTESTINAL tumors , *GLIOMAS - Abstract
The kinase PRKD2 (protein kinase D) is a crucial regulator of tumor cell-endothelial cell communication in gastrointestinal tumors and glioblastomas, but its mechanistic contributions to malignant development are not understood. Here, we report that the oncogenic chaperone HSP90 binds to and stabilizes PRKD2 in human cancer cells. Pharmacologic inhibition of HSP90 with structurally divergent small molecules currently in clinical development triggered proteasome-dependent degradation of PRKD2, augmenting apoptosis in human cancer cells of various tissue origins. Conversely, ectopic expression of PRKD2 protected cancer cells from the apoptotic effects of HSP90 abrogation, restoring blood vessel formation in two preclinical models of solid tumors. Mechanistic studies revealed that PRKD2 is essential for hypoxia-induced accumulation of hypoxia-inducible factor-1α (HIF1α) and activation of NF-κB in tumor cells. Notably, ectopic expression of PRKD2 was able to partially restore HIF1α and secreted VEGF-A levels in hypoxic cancer cells treated with HSP90 inhibitors. Taken together, our findings indicate that signals from hypoxia and HSP90 pathways are interconnected and funneled by PRKD2 into the NF-κB/VEGF-A signaling axis to promote tumor angiogenesis and tumor growth. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
21. Functional Genomic Screening in Human Pluripotent Stem Cells Reveals New Roadblocks in Early Pancreatic Endoderm Formation.
- Author
-
Krüger, Jana, Breunig, Markus, Pasquini, Lino Pascal, Morawe, Mareen, Groß, Alexander, Arnold, Frank, Russell, Ronan, Seufferlein, Thomas, Azoitei, Ninel, Kestler, Hans A., Julier, Cécile, Heller, Sandra, Hohwieler, Meike, and Kleger, Alexander
- Subjects
HUMAN stem cells ,ENDODERM ,SOMATOTYPES ,CELL analysis ,PLURIPOTENT stem cells - Abstract
Human pluripotent stem cells, with their ability to proliferate indefinitely and to differentiate into virtually all cell types of the human body, provide a novel resource to study human development and to implement relevant disease models. Here, we employed a human pancreatic differentiation platform complemented with an shRNA screen in human pluripotent stem cells (PSCs) to identify potential drivers of early endoderm and pancreatic development. Deep sequencing followed by abundancy ranking pinpointed six top hit genes potentially associated with either improved or impaired endodermal differentiation, which were selected for functional validation in CRISPR-Cas9 mediated knockout (KO) lines. Upon endoderm differentiation (DE), particularly the loss of SLC22A1 and DSC2 led to impaired differentiation efficiency into CXCR4/KIT-positive DE cells. qPCR analysis also revealed changes in differentiation markers CXCR4, FOXA2, SOX17, and GATA6. Further differentiation of PSCs to the pancreatic progenitor (PP) stage resulted in a decreased proportion of PDX1/NKX6-1-positive cells in SLC22A1 KO lines, and in DSC2 KO lines when differentiated under specific culture conditions. Taken together, our study reveals novel genes with potential roles in early endodermal development. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. Protein kinase D2 is a novel regulator of glioblastoma growth and tumor formation.
- Author
-
Azoitei, Ninel, Kleger, Alexander, Schoo, Nina, Thal, Dietmar Rudolf, Brunner, Cornelia, Pusapati, Ganesh Varma, Filatova, Alina, Genze, Felicitas, Möller, Peter, Acker, Til, Kuefer, Rainer, Van Lint, Johan, Baust, Heinrich, Adler, Guido, and Seufferlein, Thomas
- Published
- 2011
- Full Text
- View/download PDF
23. CDKN2A -Mutated Pancreatic Ductal Organoids from Induced Pluripotent Stem Cells to Model a Cancer Predisposition Syndrome.
- Author
-
Merkle, Jessica, Breunig, Markus, Schmid, Maximilian, Allgöwer, Chantal, Krüger, Jana, Melzer, Michael K., Bens, Susanne, Siebert, Reiner, Perkhofer, Lukas, Azoitei, Ninel, Seufferlein, Thomas, Heller, Sandra, Meier, Matthias, Müller, Martin, Kleger, Alexander, and Hohwieler, Meike
- Subjects
PANCREATIC tumors ,IN vitro studies ,GENETIC mutation ,PANCREATIC duct ,GENETIC disorders ,GENE expression ,TRANSITIONAL cell carcinoma ,CELL cycle ,STEM cells ,HEREDITARY cancer syndromes ,TRANSPLANTATION of organs, tissues, etc. - Abstract
Simple Summary: The mutational landscape of pancreatic ductal adenocarcinoma has been delineated in large clinical studies. Such retrospective characterization, however, is limited to late stages of the disease, and exploring the complexity of carcinogenic routes is in its infancy. Here, we provide an in vitro and in vivo model system to directly investigate human PDAC development after the induction of specific oncogenic insults in a cancer-prone ancestry. We reprogrammed plucked human hair-derived keratinocytes from two siblings harboring a pathogenic CDKN2A variant putatively predisposing for PDAC to induced pluripotent stem cells (iPSCs) and additionally introduced an inducible KRAS
G12D piggyBac expression cassette. Upon KRASG12D induction in differentiated pancreatic duct-like organoids, structural and molecular changes were observed. After orthotopic transplantation either a high-grade precancer lesion or PDAC-like tumor developed. Hereby, we provide a hereditary human pancreatic cancer model which enables further dissection of tumor initiation and early development starting from patient-specific CDKN2A-mutated pluripotent stem cells. Patient-derived induced pluripotent stem cells (iPSCs) provide a unique platform to study hereditary disorders and predisposition syndromes by resembling germline mutations of affected individuals and by their potential to differentiate into nearly every cell type of the human body. We employed plucked human hair from two siblings with a family history of cancer carrying a pathogenic CDKN2A variant, P16-p.G101W/P14-p.R115L, to generate patient-specific iPSCs in a cancer-prone ancestry for downstream analytics. The differentiation capacity to pancreatic progenitors and to pancreatic duct-like organoids (PDLOs) according to a recently developed protocol remained unaffected. Upon inducible expression of KRASG12D using a piggyBac transposon system in CDKN2A-mutated PDLOs, we revealed structural and molecular changes in vitro, including disturbed polarity and epithelial-to-mesenchymal (EMT) transition. CDKN2A-mutated KRASG12D PDLO xenotransplants formed either a high-grade precancer lesion or a partially dedifferentiated PDAC-like tumor. Intriguingly, P14/P53/P21 and P16/RB cell-cycle checkpoint controls have been only partly overcome in these grafts, thereby still restricting the tumorous growth. Hereby, we provide a model for hereditary human pancreatic cancer that enables dissection of tumor initiation and early development starting from patient-specific CDKN2A-mutated pluripotent stem cells. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
24. Telomerase and Pluripotency Factors Jointly Regulate Stemness in Pancreatic Cancer Stem Cells.
- Author
-
Walter, Karolin, Rodriguez-Aznar, Eva, Ferreira, Monica S. Ventura, Frappart, Pierre-Olivier, Dittrich, Tabea, Tiwary, Kanishka, Meessen, Sabine, Lerma, Laura, Daiss, Nora, Schulte, Lucas-Alexander, Najafova, Zeynab, Arnold, Frank, Usachov, Valentyn, Azoitei, Ninel, Erkan, Mert, Lechel, Andre, Brümmendorf, Tim H., Seufferlein, Thomas, Kleger, Alexander, and Tabarés, Enrique
- Subjects
TELOMERES ,PANCREATIC tumors ,IN vitro studies ,GENETICS ,IN vivo studies ,DNA ,TELOMERASE ,ANIMAL experimentation ,APOPTOSIS ,CANCER relapse ,STEM cells - Abstract
Simple Summary: Pancreatic ductal adenocarcinoma (PDAC) is an extremely lethal cancer with very limited therapeutic options. Cancer stem cells (CSCs) are essential for propagation of PDAC, but also for its metastatic activity and chemoresistance. To date, it is still unclear how cancer stem cells (CSCs) regulate their 'stemness' and self-renewal properties, and to what extent they share common features with normal stem cells. Telomerase regulation is a key factor in stem cell maintenance. Here, we investigate how telomerase regulation affects CSC biology in PDAC, and delineate the mechanisms by which telomerase activity and CSC properties are linked. To assess the role of telomerase activity and telomere length in pancreatic CSCs we used different CSC enrichment methods (CD133, ALDH, sphere formation) in primary patient-derived pancreatic cancer cells. We show that CSCs have higher telomerase activity and longer telomeres than bulk tumor cells. Inhibition of telomerase activity, using genetic knockdown or pharmacological inhibitor (BIBR1532), resulted in CSC marker depletion, abrogation of sphere formation in vitro and reduced tumorigenicity in vivo. Furthermore, we identify a positive feedback loop between stemness factors (NANOG, OCT3/4, SOX2, KLF4) and telomerase, which is essential for the self-renewal of CSCs. Disruption of the balance between telomerase activity and stemness factors eliminates CSCs via induction of DNA damage and apoptosis in primary patient-derived pancreatic cancer samples, opening future perspectives to avoid CSC-driven tumor relapse. In the present study, we demonstrate that telomerase regulation is critical for the "stemness" maintenance in pancreatic CSCs and examine the effects of telomerase inhibition as a potential treatment option of pancreatic cancer. This may significantly promote our understanding of PDAC tumor biology and may result in improved treatment for pancreatic cancer patients. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. An Immunological Glance on Pancreatic Ductal Adenocarcinoma.
- Author
-
Melzer, Michael Karl, Arnold, Frank, Stifter, Katja, Zengerling, Friedemann, Azoitei, Ninel, Seufferlein, Thomas, Bolenz, Christian, and Kleger, Alexander
- Subjects
PANCREATIC cancer ,ADENOCARCINOMA ,CANCER cells ,IMMUNE system ,THERAPEUTICS ,GLEASON grading system - Abstract
Pancreatic ductal adenocarcinoma (PDAC) has still a dismal prognosis. Different factors such as mutational landscape, intra- and intertumoral heterogeneity, stroma, and immune cells impact carcinogenesis of PDAC associated with an immunosuppressive microenvironment. Different cell types with partly opposing roles contribute to this milieu. In recent years, immunotherapeutic approaches, including checkpoint inhibitors, were favored to treat cancers, albeit not every cancer entity exhibited benefits in a similar way. Indeed, immunotherapies rendered little success in pancreatic cancer. In this review, we describe the communication between the immune system and pancreatic cancer cells and propose some rationale why immunotherapies may fail in the context of pancreatic cancer. Moreover, we delineate putative strategies to sensitize PDAC towards immunological therapeutics and highlight the potential of targeting neoantigens. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
26. A time frame permissive for Protein Kinase D2 activity to direct angiogenesis in mouse embryonic stem cells.
- Author
-
Müller, Martin, Schröer, Jana, Azoitei, Ninel, Eiseler, Tim, Bergmann, Wendy, Köhntop, Ralf, Weidgang, Clair, Seufferlein, Thomas, Kleger, Alexander, Lin, Qiong, Zenke, Martin, Costa, Ivan G, Genze, Felicitas, and Liebau, Stefan
- Subjects
PROTEIN kinases ,ISOENZYMES ,PHOSPHOLIPASE D ,BIOLOGICAL systems ,NEOVASCULARIZATION inhibitors - Abstract
The protein kinase D isoenzymes PKD1/2/3 are prominent downstream targets of PKCs (Protein Kinase Cs) and phospholipase D in various biological systems. Recently, we identified PKD isoforms as novel mediators of tumour cell-endothelial cell communication, tumour cell motility and metastasis. Although PKD isoforms have been implicated in physiological/tumour angiogenesis, a role of PKDs during embryonic development, vasculogenesis and angiogenesis still remains elusive. We investigated the role of PKDs in germ layer segregation and subsequent vasculogenesis and angiogenesis using mouse embryonic stem cells (ESCs). We show that mouse ESCs predominantly express PKD2 followed by PKD3 while PKD1 displays negligible levels. Furthermore, we demonstrate that PKD2 is specifically phosphorylated/activated at the time of germ layer segregation. Time-restricted PKD2-activation limits mesendoderm formation and subsequent cardiovasculogenesis during early differentiation while leading to branching angiogenesis during late differentiation. In line, PKD2 loss-of-function analyses showed induction of mesendodermal differentiation in expense of the neuroectodermal germ layer. Our in vivo findings demonstrate that embryoid bodies transplanted on chicken chorioallantoic membrane induced an angiogenic response indicating that timed overexpression of PKD2 from day 4 onwards leads to augmented angiogenesis in differentiating ESCs. Taken together, our results describe novel and time-dependent facets of PKD2 during early cell fate determination. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
27. Thirty-eight-negative kinase 1 mediates trauma-induced intestinal injury and multi-organ failure.
- Author
-
Armacki, Milena, Trugenberger, Anna Katharina, Ellwanger, Ann K., Eiseler, Tim, Schwerdt, Christiane, Bettac, Lucas, Langgartner, Dominik, Azoitei, Ninel, Halbgebauer, Rebecca, Groß, Rüdiger, Barth, Tabea, Lechel, André, Walter, Benjamin M., Kraus, Johann M., Wiegreffe, Christoph, Grimm, Johannes, Scheffold, Annika, Schneider, Marlon R., Peuker, Kenneth, and Zeißig, Sebastian
- Subjects
- *
EPITHELIAL cells , *APOPTOSIS , *SYSTEMIC inflammatory response syndrome , *GASTROINTESTINAL diseases , *KINASES , *COLITIS - Abstract
Dysregulated intestinal epithelial apoptosis initiates gut injury, alters the intestinal barrier, and can facilitate bacterial translocation leading to a systemic inflammatory response syndrome (SIRS) and/or multi-organ dysfunction syndrome (MODS). A variety of gastrointestinal disorders, including inflammatory bowel disease, have been linked to intestinal apoptosis. Similarly, intestinal hyperpermeability and gut failure occur in critically ill patients, putting the gut at the center of SIRS pathology. Regulation of apoptosis and immune-modulatory functions have been ascribed to Thirty-eight-negative kinase 1 (TNK1), whose activity is regulated merely by expression. We investigated the effect of TNK1 on intestinal integrity and its role in MODS. TNK1 expression induced crypt-specific apoptosis, leading to bacterial translocation, subsequent septic shock, and early death. Mechanistically, TNK1 expression in vivo resulted in STAT3 phosphorylation, nuclear translocation of p65, and release of IL-6 and TNF-α. A TNF-α neutralizing antibody partially blocked development of intestinal damage. Conversely, gut-specific deletion of TNK1 protected the intestinal mucosa from experimental colitis and prevented cytokine release in the gut. Finally, TNK1 was found to be deregulated in the gut in murine and porcine trauma models and human inflammatory bowel disease. Thus, TNK1 might be a target during MODS to prevent damage in several organs, notably the gut. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
28. Overexpression of Phospholipid-hydroperoxide Glutathione Peroxidase in Human Dermal Fibroblasts Abrogates UVA Irradiation-induced Expression of Interstitial Collagenase/Matrix Metalloproteinase-1...
- Author
-
Wenk, Jutta, Schüller, Jutta, Hinrichs, Christina, Syrovets, Tatjana, Azoitei, Ninel, Podda, Maurizio, Wlaschek, Meinhard, Brenneisen, Peter, Schneider, Lars-A., Sabiwalsky, Andrea, Peters, Thorsten, Sulyok, Silke, Dissemond, Joachim, Schauen, Matthias, Krieg, Thomas, Wirth, Thomas, Simmet, Thomas, and Scharffetter-Kochanek, Karin
- Subjects
- *
PEROXIDASE , *METALLOENZYMES , *PHOSPHOLIPIDS , *GLUTATHIONE , *FIBROBLASTS , *ULTRAVIOLET radiation , *COLLAGENASES , *METALLOPROTEINASES - Abstract
Phospholipid-hydroperoxide glutathione peroxidase (PHGPx) exhibits high specific activity in reducing phosphatidylcholine hydroperoxides (PCOOHs) and thus may play a central role in protecting the skin against UV irradiation-triggered detrimental long term effects like cancer formation and premature skin aging. Here we addressed the role of PHGPx in the protection against UV irradiation-induced expression of matrix metalloproteinase-1 (MMP-1). For this purpose, we created human dermal fibroblast cell lines overexpressing human PHGPx. Overexpression led to a significant increase in PHGPx activity. In contrast to a maximal 4.5-fold induction of specific MMP-1 mRNA levels in vector-transfected cells at 24 h after UVA irradiation, no MMP-1 induction occurred at any studied time point after UVA treatment of PHGPx-overexpressing fibroblasts. As interleukin-6 (IL-6) was earlier shown to mediate the UVA induction of MMP-1, we studied whether PHGPx overexpression might interfere with the NFκB-mediated IL-6 induction and downstream signaling. Using transient transfections of IL-6 promoter constructs containing NFκB binding sites, we observed a high induction of the reporter gene luciferase in vector-transfected control cells and a significantly lower induction in PHGPx-overexpressing fibroblasts following UVA irradiation. Consistently both UVA irradiation and treatment of fibroblasts with PCOOHs led to phosphorylation and nuclear translocation of the p65 subunit, whereas cells overexpressing PHGPx exhibited impaired NFκB activation, p65 phosphorylation, and nuclear translocation. In line with this, the PHGPx-overexpressing fibroblasts [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
29. Modeling plasticity and dysplasia of pancreatic ductal organoids derived from human pluripotent stem cells.
- Author
-
Breunig M, Merkle J, Wagner M, Melzer MK, Barth TFE, Engleitner T, Krumm J, Wiedenmann S, Cohrs CM, Perkhofer L, Jain G, Krüger J, Hermann PC, Schmid M, Madácsy T, Varga Á, Griger J, Azoitei N, Müller M, Wessely O, Robey PG, Heller S, Dantes Z, Reichert M, Günes C, Bolenz C, Kuhn F, Maléth J, Speier S, Liebau S, Sipos B, Kuster B, Seufferlein T, Rad R, Meier M, Hohwieler M, and Kleger A
- Subjects
- Animals, Humans, Mice, Mutation, Organoids, Pancreatic Ducts, Proteomics, Carcinoma, Pancreatic Ductal, Pancreatic Neoplasms genetics, Pluripotent Stem Cells
- Abstract
Personalized in vitro models for dysplasia and carcinogenesis in the pancreas have been constrained by insufficient differentiation of human pluripotent stem cells (hPSCs) into the exocrine pancreatic lineage. Here, we differentiate hPSCs into pancreatic duct-like organoids (PDLOs) with morphological, transcriptional, proteomic, and functional characteristics of human pancreatic ducts, further maturing upon transplantation into mice. PDLOs are generated from hPSCs inducibly expressing oncogenic GNAS, KRAS, or KRAS with genetic covariance of lost CDKN2A and from induced hPSCs derived from a McCune-Albright patient. Each oncogene causes a specific growth, structural, and molecular phenotype in vitro. While transplanted PDLOs with oncogenic KRAS alone form heterogenous dysplastic lesions or cancer, KRAS with CDKN2A loss develop dedifferentiated pancreatic ductal adenocarcinomas. In contrast, transplanted PDLOs with mutant GNAS lead to intraductal papillary mucinous neoplasia-like structures. Conclusively, PDLOs enable in vitro and in vivo studies of pancreatic plasticity, dysplasia, and cancer formation from a genetically defined background., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. RINT1 Regulates SUMOylation and the DNA Damage Response to Preserve Cellular Homeostasis in Pancreatic Cancer.
- Author
-
Arnold F, Gout J, Wiese H, Weissinger SE, Roger E, Perkhofer L, Walter K, Scheible J, Prelli Bozzo C, Lechel A, Ettrich TJ, Azoitei N, Hao L, Fürstberger A, Kaminska EK, Sparrer KMJ, Rasche V, Wiese S, Kestler HA, Möller P, Seufferlein T, Frappart PO, and Kleger A
- Subjects
- Animals, Cell Line, Tumor, Cohort Studies, DNA Damage genetics, Female, Homeostasis genetics, Humans, Mice, Mice, Nude, Mice, Transgenic, Protein Processing, Post-Translational genetics, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Cycle Proteins physiology, DNA Repair genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Sumoylation genetics
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) still presents with a dismal prognosis despite intense research. Better understanding of cellular homeostasis could identify druggable targets to improve therapy. Here we propose RAD50-interacting protein 1 (RINT1) as an essential mediator of cellular homeostasis in PDAC. In a cohort of resected PDAC, low RINT1 protein expression correlated significantly with better survival. Accordingly, RINT1 depletion caused severe growth defects in vitro associated with accumulation of DNA double-strand breaks (DSB), G
2 cell cycle arrest, disruption of Golgi-endoplasmic reticulum homeostasis, and cell death. Time-resolved transcriptomics corroborated by quantitative proteome and interactome analyses pointed toward defective SUMOylation after RINT1 loss, impairing nucleocytoplasmic transport and DSB response. Subcutaneous xenografts confirmed tumor response by RINT1 depletion, also resulting in a survival benefit when transferred to an orthotopic model. Primary human PDAC organoids licensed RINT1 relevance for cell viability. Taken together, our data indicate that RINT1 loss affects PDAC cell fate by disturbing SUMOylation pathways. Therefore, a RINT1 interference strategy may represent a new putative therapeutic approach. SIGNIFICANCE: These findings provide new insights into the aggressive behavior of PDAC, showing that RINT1 directly correlates with survival in patients with PDAC by disturbing the SUMOylation process, a crucial modification in carcinogenesis., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
31. Synergistic targeting and resistance to PARP inhibition in DNA damage repair-deficient pancreatic cancer.
- Author
-
Gout J, Perkhofer L, Morawe M, Arnold F, Ihle M, Biber S, Lange S, Roger E, Kraus JM, Stifter K, Hahn SA, Zamperone A, Engleitner T, Müller M, Walter K, Rodriguez-Aznar E, Sainz B Jr, Hermann PC, Hessmann E, Müller S, Azoitei N, Lechel A, Liebau S, Wagner M, Simeone DM, Kestler HA, Seufferlein T, Wiesmüller L, Rad R, Frappart PO, and Kleger A
- Subjects
- Adenocarcinoma drug therapy, Animals, Apoptosis, Carcinoma, Pancreatic Ductal drug therapy, Cell Line, Tumor, Cell Survival, DNA Copy Number Variations, DNA Damage, DNA Repair, Drug Resistance, Multiple genetics, Drug Synergism, Epithelial-Mesenchymal Transition, Genotype, Humans, Mice, Pancreatic Neoplasms drug therapy, Prognosis, Adenocarcinoma genetics, Ataxia Telangiectasia Mutated Proteins genetics, Carcinoma, Pancreatic Ductal genetics, Homologous Recombination, Pancreatic Neoplasms genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology
- Abstract
Objective: ATM serine/threonine kinase (ATM) is the most frequently mutated DNA damage response gene, involved in homologous recombination (HR), in pancreatic ductal adenocarcinoma (PDAC)., Design: Combinational synergy screening was performed to endeavour a genotype-tailored targeted therapy., Results: Synergy was found on inhibition of PARP, ATR and DNA-PKcs (PAD) leading to synthetic lethality in ATM-deficient murine and human PDAC. Mechanistically, PAD-induced PARP trapping, replication fork stalling and mitosis defects leading to P53-mediated apoptosis. Most importantly, chemical inhibition of ATM sensitises human PDAC cells toward PAD with long-term tumour control in vivo. Finally, we anticipated and elucidated PARP inhibitor resistance within the ATM-null background via whole exome sequencing. Arising cells were aneuploid, underwent epithelial-mesenchymal-transition and acquired multidrug resistance (MDR) due to upregulation of drug transporters and a bypass within the DNA repair machinery. These functional observations were mirrored in copy number variations affecting a region on chromosome 5 comprising several of the upregulated MDR genes. Using these findings, we ultimately propose alternative strategies to overcome the resistance., Conclusion: Analysis of the molecular susceptibilities triggered by ATM deficiency in PDAC allow elaboration of an efficient mutation-specific combinational therapeutic approach that can be also implemented in a genotype-independent manner by ATM inhibition., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
32. Variant classification in precision oncology.
- Author
-
Leichsenring J, Horak P, Kreutzfeldt S, Heining C, Christopoulos P, Volckmar AL, Neumann O, Kirchner M, Ploeger C, Budczies J, Heilig CE, Hutter B, Fröhlich M, Uhrig S, Kazdal D, Allgäuer M, Harms A, Rempel E, Lehmann U, Thomas M, Pfarr N, Azoitei N, Bonzheim I, Marienfeld R, Möller P, Werner M, Fend F, Boerries M, von Bubnoff N, Lassmann S, Longerich T, Bitzer M, Seufferlein T, Malek N, Weichert W, Schirmacher P, Penzel R, Endris V, Brors B, Klauschen F, Glimm H, Fröhling S, and Stenzinger A
- Subjects
- Humans, Molecular Targeted Therapy, Mutation, Neoplasms drug therapy, Precision Medicine, Sequence Analysis, DNA, High-Throughput Nucleotide Sequencing methods, Neoplasms genetics
- Abstract
Next-generation sequencing has become a cornerstone of therapy guidance in cancer precision medicine and an indispensable research tool in translational oncology. Its rapidly increasing use during the last decade has expanded the options for targeted tumor therapies, and molecular tumor boards have grown accordingly. However, with increasing detection of genetic alterations, their interpretation has become more complex and error-prone, potentially introducing biases and reducing benefits in clinical practice. To facilitate interdisciplinary discussions of genetic alterations for treatment stratification between pathologists, oncologists, bioinformaticians, genetic counselors and medical scientists in specialized molecular tumor boards, several systems for the classification of variants detected by large-scale sequencing have been proposed. We review three recent and commonly applied classifications and discuss their individual strengths and weaknesses. Comparison of the classifications underlines the need for a clinically useful and universally applicable variant reporting system, which will be instrumental for efficient decision making based on sequencing analysis in oncology. Integrating these data, we propose a generalizable classification concept featuring a conservative and a more progressive scheme, which can be readily applied in a clinical setting., (© 2019 UICC.)
- Published
- 2019
- Full Text
- View/download PDF
33. ATM Deficiency Generating Genomic Instability Sensitizes Pancreatic Ductal Adenocarcinoma Cells to Therapy-Induced DNA Damage.
- Author
-
Perkhofer L, Schmitt A, Romero Carrasco MC, Ihle M, Hampp S, Ruess DA, Hessmann E, Russell R, Lechel A, Azoitei N, Lin Q, Liebau S, Hohwieler M, Bohnenberger H, Lesina M, Algül H, Gieldon L, Schröck E, Gaedcke J, Wagner M, Wiesmüller L, Sipos B, Seufferlein T, Reinhardt HC, Frappart PO, and Kleger A
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Fluorouracil pharmacology, Gene Expression, Genomic Instability, Humans, Immunohistochemistry, Male, Mice, Mice, SCID, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Phthalazines pharmacology, Piperazines pharmacology, Gemcitabine, Ataxia Telangiectasia Mutated Proteins deficiency, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal enzymology, DNA Damage, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms enzymology
- Abstract
Pancreatic ductal adenocarcinomas (PDAC) harbor recurrent functional mutations of the master DNA damage response kinase ATM, which has been shown to accelerate tumorigenesis and epithelial-mesenchymal transition. To study how ATM deficiency affects genome integrity in this setting, we evaluated the molecular and functional effects of conditional Atm deletion in a mouse model of PDAC. ATM deficiency was associated with increased mitotic defects, recurrent genomic rearrangements, and deregulated DNA integrity checkpoints, reminiscent of human PDAC. We hypothesized that altered genome integrity might allow synthetic lethality-based options for targeted therapeutic intervention. Supporting this possibility, we found that the PARP inhibitor olaparib or ATR inhibitors reduced the viability of PDAC cells in vitro and in vivo associated with a genotype-selective increase in apoptosis. Overall, our results offered a preclinical mechanistic rationale for the use of PARP and ATR inhibitors to improve treatment of ATM-mutant PDAC. Cancer Res; 77(20); 5576-90. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
34. STK33 participates to HSP90-supported angiogenic program in hypoxic tumors by regulating HIF-1α/VEGF signaling pathway.
- Author
-
Liu Y, Steinestel K, Rouhi A, Armacki M, Diepold K, Chiosis G, Simmet T, Seufferlein T, and Azoitei N
- Abstract
Lately, the HSP90 client serine/threonine kinase STK33 emerged to be required by cancer cells for their viability and proliferation. However, its mechanistic contribution to carcinogenesis is not clearly understood. Here we report that elevated STK33 expression correlates with advanced stages of human pancreatic and colorectal carcinomas. Impaired proliferation and augmented apoptosis associated with genetic abrogation of STK33 were paralleled by decreased vascularization in tumor xenografts. In line with this, ectopic STK33 not only promoted tumor growth after pharmacologic inhibition of HSP90 using structurally divergent small molecules currently in clinical development, but also restored blood vessel formation in vivo . Mechanistic studies demonstrated that HSP90-stabilized STK33 interacts with and regulates hypoxia-driven accumulation and activation of HIF-1α as well as secretion of VEGF-A in hypoxic cancer cells. In addition, our study reveals a putative cooperation between STK33 and other HSP90 client protein kinases involved in molecular and cellular events through which cancer cells ensure their survival by securing the oxygen and nutrient supply. Altogether, our findings indicate that STK33 interferes with signals from hypoxia and HSP90 to promote tumor angiogenesis and tumor growth., Competing Interests: CONFLICTS OF INTEREST Memorial Sloan-Kettering Cancer Center holds the intellectual rights to PU-H71. Samus Therapeutics, of which G. Chiosis has partial ownership, has licensed PU-H71.
- Published
- 2017
- Full Text
- View/download PDF
35. PRKD2: A two-pronged kinase crucial for the tumor-supporting activity of HSP90.
- Author
-
Azoitei N, Fröhling S, Scholl C, and Seufferlein T
- Abstract
PRKD2 plays an important role in tumor cell survival, proliferation, migration, and angiogenesis. We recently reported that cell death and impaired blood vessel formation evoked by inhibition of the HSP90 chaperone in human cancer cells of various tissue origins is mediated by destabilization of PRKD2.
- Published
- 2015
- Full Text
- View/download PDF
36. Targeting of KRAS mutant tumors by HSP90 inhibitors involves degradation of STK33.
- Author
-
Azoitei N, Hoffmann CM, Ellegast JM, Ball CR, Obermayer K, Gößele U, Koch B, Faber K, Genze F, Schrader M, Kestler HA, Döhner H, Chiosis G, Glimm H, Fröhling S, and Scholl C
- Subjects
- Apoptosis, Cell Line, Tumor, Colonic Neoplasms drug therapy, Colonic Neoplasms pathology, Humans, Proteasome Endopeptidase Complex physiology, Proto-Oncogene Proteins physiology, Proto-Oncogene Proteins p21(ras), Ubiquitination, ras Proteins physiology, HSP90 Heat-Shock Proteins antagonists & inhibitors, Mutation, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins genetics, ras Proteins genetics
- Abstract
Previous efforts to develop drugs that directly inhibit the activity of mutant KRAS, the most commonly mutated human oncogene, have not been successful. Cancer cells driven by mutant KRAS require expression of the serine/threonine kinase STK33 for their viability and proliferation, identifying STK33 as a context-dependent therapeutic target. However, specific strategies for interfering with the critical functions of STK33 are not yet available. Here, using a mass spectrometry-based screen for STK33 protein interaction partners, we report that the HSP90/CDC37 chaperone complex binds to and stabilizes STK33 in human cancer cells. Pharmacologic inhibition of HSP90, using structurally divergent small molecules currently in clinical development, induced proteasome-mediated degradation of STK33 in human cancer cells of various tissue origin in vitro and in vivo, and triggered apoptosis preferentially in KRAS mutant cells in an STK33-dependent manner. Furthermore, HSP90 inhibitor treatment impaired sphere formation and viability of primary human colon tumor-initiating cells harboring mutant KRAS. These findings provide mechanistic insight into the activity of HSP90 inhibitors in KRAS mutant cancer cells, indicate that the enhanced requirement for STK33 can be exploited to target mutant KRAS-driven tumors, and identify STK33 depletion through HSP90 inhibition as a biomarker-guided therapeutic strategy with immediate translational potential.
- Published
- 2012
- Full Text
- View/download PDF
37. NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression.
- Author
-
Huber MA, Azoitei N, Baumann B, Grünert S, Sommer A, Pehamberger H, Kraut N, Beug H, and Wirth T
- Subjects
- Animals, Apoptosis drug effects, Blotting, Western, Breast Neoplasms, Cell Line, Transformed, Cell Line, Tumor, Cell Transformation, Neoplastic chemically induced, Cell Transformation, Neoplastic drug effects, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Cell Transformation, Viral, Disease Models, Animal, Epithelial Cells metabolism, Epithelial Cells transplantation, Gene Expression Regulation, Neoplastic, Immunohistochemistry, Mammary Glands, Animal drug effects, Mammary Glands, Animal metabolism, Mammary Glands, Animal pathology, Mesoderm drug effects, Mesoderm metabolism, Mice, Mice, Nude, NF-kappa B antagonists & inhibitors, NF-kappa B genetics, Neoplasm Transplantation, Oncogene Protein p21(ras) genetics, Oncogene Protein p21(ras) metabolism, Retroviridae genetics, Time Factors, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta pharmacology, Cell Transformation, Neoplastic genetics, Epithelial Cells drug effects, Epithelial Cells pathology, NF-kappa B metabolism, Neoplasm Metastasis
- Abstract
The transcription factor NF-kappaB is activated in a range of human cancers and is thought to promote tumorigenesis, mainly due to its ability to protect transformed cells from apoptosis. To investigate the role of NF-kappaB in epithelial plasticity and metastasis, we utilized a well-characterized in vitro/in vivo model of mammary carcinogenesis that depends on the collaboration of the Ha-Ras oncoprotein and TGF-beta. We show here that the IKK-2/IkappaBalpha/NF-kappaB pathway is required for the induction and maintenance of epithelial-mesenchymal transition (EMT). Inhibition of NF-kappaB signaling prevented EMT in Ras-transformed epithelial cells, while activation of this pathway promoted the transition to a mesenchymal phenotype even in the absence of TGF-beta. Furthermore, inhibition of NF-kappaB activity in mesenchymal cells caused a reversal of EMT, suggesting that NF-kappaB is essential for both the induction and maintenance of EMT. In line with the importance of EMT for invasion, blocking of NF-kappaB activity abrogated the metastatic potential of mammary epithelial cells in a mouse model system. Collectively, these data provide evidence of an essential role for NF-kappaB during distinct steps of breast cancer progression and suggest that the cooperation of Ras- and TGF-beta-dependent signaling pathways in late-stage tumorigenesis depends critically on NF-kappaB activity.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.