90 results on '"Schramek, D."'
Search Results
2. RANK und RANKL - Vom Knochen zum Mammakarzinom
- Author
-
Sigl V, Schramek D, and Penninger JM
- Subjects
Stammzellen ,Progesteron ,Knochen ,lcsh:R ,lcsh:Medicine ,Onkologie - Abstract
RANK (Receptor Activator of NF-κB) und sein Ligand RANKL sind Schlüsselmoleküle im Knochenmetabolismus und spielen eine essenzielle Rolle in der Entstehung von pathologischen Knochenveränderungen. Die Deregulation des RANK/RANKL-Systems ist zum Beispiel ein Hauptgrund für das Auftreten von postmenopausaler Osteoporose bei Frauen. Eine weitere wesentliche Funktion von RANK und RANKL liegt in der Entwicklung von milchsekretierenden Drüsen während der Schwangerschaft. Dabei regulieren Sexualhormone, wie zum Beispiel Progesteron, die Expression von RANKL und induzieren dadurch die Proliferation von epithelialen Zellen der Brust. Seit Längerem war schon bekannt, dass RANK und RANKL in der Metastasenbildung von Brustkrebszellen im Knochengewebe beteiligt sind. Wir konnten nun das RANK/RANKLSystem auch als essenziellen Mechanismus in der Entstehung von hormonellem Brustkrebs identifizieren. In diesem Beitrag werden wir daher den neuesten Erkenntnissen besondere Aufmerksamkeit schenken und diese kritisch in Bezug auf Brustkrebsentwicklung betrachten.
- Published
- 2012
3. MKK7 and ARF: New players in the DNA damage response scenery
- Author
-
Kotsinas, A. Papanagnou, P. Galanos, P. Schramek, D. Townsend, P. Penninger, J.M. Bartek, J. Gorgoulis, V.G.
- Subjects
body regions - Abstract
Sensing, integrating, and processing of stressogenic signals must be followed by accurate differential response(s) for a cell to survive and avoid malignant transformation. The DNA damage response (DDR) pathway is vital in this process, as it deals with genotoxic/oncogenic insults, having p53 as a nodal effector that performs most of the above tasks. Accumulating data reveal that other pathways are also involved in the same or similar processes, conveying also to p53. Emerging questions are if, how, and when these additional pathways communicate with the DDR axis. Two such stress response pathways, involving the MKK7 stress-activated protein kinase (SAPK) and ARF, have been shown to be interlocked with the ATM/ATR-regulated DDR axis in a highly ordered manner. This creates a new landscape in the DDR orchestrated response to genotoxic/oncogenic insults that is currently discussed. © 2014 Landes Bioscience.
- Published
- 2014
4. A Multi-Hit mouse model to identify cooperating Ras effector pathways in lung cancer
- Author
-
Musteanu M, Blaas L, Zenz R, Svinka J, Hoffmann T, Grabner B, Schramek D, Kantner HP, Mxfcller M, Kolbe T, Rxfclicke T, Moriggl R, Kenner L, Stoiber D, Penninger JM, Popper H, Casanova E, and Eferl R
- Published
- 2012
5. The stress kinase MKK7 couples oncogenic stress to p53 stability and tumor suppression
- Author
-
Schramek, D. Kotsinas, A. Meixner, A. Wada, T. Elling, U. Pospisilik, J.A. Neely, G.G. Zwick, R.-H. Sigl, V. Forni, G. Serrano, M. Gorgoulis, V.G. Penninger, J.M.
- Abstract
Most preneoplastic lesions are quiescent and do not progress to form overt tumors. It has been proposed that oncogenic stress activates the DNA damage response and the key tumor suppressor p53, which prohibits tumor growth. However, the molecular pathways by which cells sense a premalignant state in vivo are largely unknown. Here we report that tissue-specific inactivation of the stress signaling kinase MKK7 in KRas G12D-driven lung carcinomas and NeuT-driven mammary tumors markedly accelerates tumor onset and reduces overall survival. Mechanistically, MKK7 acts through the kinases JNK1 and JNK2, and this signaling pathway directly couples oncogenic and genotoxic stress to the stability of p53, which is required for cell cycle arrest and suppression of epithelial cancers. These results show that MKK7 functions as a major tumor suppressor in lung and mammary cancer in mouse and identify MKK7 as a vital molecular sensor to set a cellular anti-cancer barrier. © 2011 Nature America, Inc. All rights reserved.
- Published
- 2011
6. Genome-Wide RNAi Screen Identifies Genes Involved in Intestinal Pathogenic Bacterial Infection
- Author
-
Cronin SJ, Nehme NT, Limmer S, Liegeois S, Pospisilik JA, Schramek D, Leibbrandt A, Simoes Rde M, Gruber S, Puc U, Ebersberger I, Zoranovic T, Neely GG, von Haeseler A, Ferrandon D, and Penninger JM
- Published
- 2009
7. Progesterone drives mammary secretory differentiation via RankL-mediated induction of Elf5 in luminal progenitor cells.
- Author
-
Lee, HJ, Gallego-Ortega, D, Ledger, A, Schramek, D, Joshi, P, Szwarc, MM, Cho, C, Lydon, JP, Khokha, R, Penninger, JM, Ormandy, CJ, Lee, HJ, Gallego-Ortega, D, Ledger, A, Schramek, D, Joshi, P, Szwarc, MM, Cho, C, Lydon, JP, Khokha, R, Penninger, JM, and Ormandy, CJ
- Abstract
Progesterone-RankL paracrine signaling has been proposed as a driver of stem cell expansion in the mammary gland, and Elf5 is essential for the differentiation of mammary epithelial progenitor cells. We demonstrate that Elf5 expression is induced by progesterone and that Elf5 and progesterone cooperate to promote alveolar development. The progesterone receptor and Elf5 are expressed in a mutually exclusive pattern, and we identify RankL as the paracrine mediator of the effects of progesterone on Elf5 expression in CD61+ progenitor cells and their consequent differentiation. Blockade of RankL action prevented progesterone-induced side branching and the expansion of Elf5(+) mature luminal cells. These findings describe a mechanism by which steroid hormones can produce the expansion of steroid hormone receptor-negative mammary epithelial cells.
- Published
- 2013
8. Gab2 signaling in chronic myeloid leukemia cells confers resistance to multiple Bcr-Abl inhibitors
- Author
-
Wöhrle, F U, primary, Halbach, S, additional, Aumann, K, additional, Schwemmers, S, additional, Braun, S, additional, Auberger, P, additional, Schramek, D, additional, Penninger, J M, additional, Laßmann, S, additional, Werner, M, additional, Waller, C F, additional, Pahl, H L, additional, Zeiser, R, additional, Daly, R J, additional, and Brummer, T, additional
- Published
- 2012
- Full Text
- View/download PDF
9. 1050 THE APELIN/APJ SYSTEM LINKS ANGIOGENESIS TO LIVER FIBROSIS
- Author
-
Oesterreicher, C., primary, Schramek, D., additional, Penz-Österreicher, M., additional, Kuba, K., additional, Penninger, J.M., additional, and Brenner, D., additional
- Published
- 2011
- Full Text
- View/download PDF
10. 277 ROLE OF STAT5 IN LIVER FIBROSIS
- Author
-
Blaas, L., primary, Kornfeld, J.-W., additional, Zollner, G., additional, Schramek, D., additional, Esterbauer, H., additional, Trauner, M., additional, and Casanova, E., additional
- Published
- 2009
- Full Text
- View/download PDF
11. The docking protein and proto-oncogene product Gab2 is regulated via a novel negative feedback mechanism mediated by 14-3-3 binding
- Author
-
Brummer, T, primary, Larance, M, additional, Abreu, MT Herrera, additional, Lyons, RJ, additional, Timpson, P, additional, Emmerich, CH, additional, Fleuren, EDG, additional, Lehrbach, GM, additional, Schramek, D, additional, Guilhaus, M, additional, James, DE, additional, and Daly, RJ, additional
- Published
- 2009
- Full Text
- View/download PDF
12. Metabolic Regulation of the Epigenome Drives Lethal Infantile Ependymoma
- Author
-
Samuel Weiss, Leo J.Y. Kim, Xiaochong Wu, Randy Van Ommeren, Yanqing Jiang, Kaitlin Kharas, Evgeny Kanshin, Moloud Ahmadi, Alberto Delaidelli, Geneviève Deblois, David Przelicki, Stephane Angers, Hiromichi Suzuki, Sameer Agnihotri, Bradly G. Wouters, Graham MacLeod, Ricky Tsai, Pasqualino De Antonellis, Michelle Ly, Stacey L. Krumholtz, Paul Guilhamon, James Loukides, Ravi N. Vellanki, Alex Rasnitsyn, Hamza Farooq, Daniel Schramek, Nada Jabado, María Sánchez-Osuna, Laura K. Donovan, Vijay Ramaswamy, Ibrahim El-Hamamy, Joonas Haapasalo, Jeremy N. Rich, Michael D. Taylor, Benjamin A. Garcia, Mike Tyers, Kyle Juraschka, Winnie Ong, Olivier Saulnier, Panagiotis Prinos, John J.Y. Lee, Borja L. Holgado, Olga Sirbu, Craig Daniels, Cheryl H. Arrowsmith, Cory Richman, Poul H. Sorensen, Kulandaimanuvel Antony Michealraj, Sheila K. Singh, Andrea Bajic, Polina Balin, Stephen C. Mack, Betty Luu, Fiona J. Coutinho, Dilakshan Srikanthan, Florence M.G. Cavalli, Sachin Kumar, Evan Y. Wang, Mathieu Lupien, Peter B. Dirks, Maria C. Vladoiu, Lincoln Stein, Livia Garzia, Ahmad Malik, John Wojcik, Avesta Rastan, Michealraj, K. A., Kumar, S. A., Kim, L. J. Y., Cavalli, F. M. G., Przelicki, D., Wojcik, J. B., Delaidelli, A., Bajic, A., Saulnier, O., Macleod, G., Vellanki, R. N., Vladoiu, M. C., Guilhamon, P., Ong, W., Lee, J. J. Y., Jiang, Y., Holgado, B. L., Rasnitsyn, A., Malik, A. A., Tsai, R., Richman, C. M., Juraschka, K., Haapasalo, J., Wang, E. Y., De Antonellis, P., Suzuki, H., Farooq, H., Balin, P., Kharas, K., Van Ommeren, R., Sirbu, O., Rastan, A., Krumholtz, S. L., Ly, M., Ahmadi, M., Deblois, G., Srikanthan, D., Luu, B., Loukides, J., Wu, X., Garzia, L., Ramaswamy, V., Kanshin, E., Sanchez-Osuna, M., El-Hamamy, I., Coutinho, F. J., Prinos, P., Singh, S., Donovan, L. K., Daniels, C., Schramek, D., Tyers, M., Weiss, S., Stein, L. D., Lupien, M., Wouters, B. G., Garcia, B. A., Arrowsmith, C. H., Sorensen, P. H., Angers, S., Jabado, N., Dirks, P. B., Mack, S. C., Agnihotri, S., Rich, J. N., and Taylor, M. D.
- Subjects
Epigenomics ,Ependymoma ,Male ,ependymoma ,Epigenomic ,Somatic cell ,cancer metabolism ,Infratentorial Neoplasms ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Histones ,Brain Neoplasm ,03 medical and health sciences ,Epigenome ,0302 clinical medicine ,Histone demethylation ,Histone methylation ,medicine ,Animals ,Humans ,Epigenetics ,030304 developmental biology ,hindbrain development ,Cell Proliferation ,Infratentorial Neoplasm ,0303 health sciences ,Brain Neoplasms ,Animal ,Lysine ,Infant ,DNA Methylation ,medicine.disease ,microenvironment ,Mice, Inbred C57BL ,Histone ,Acetylation ,paediatric cancer ,Mutation ,biology.protein ,Cancer research ,030217 neurology & neurosurgery ,epigenetic ,Human - Abstract
Posterior fossa A (PFA) ependymomas are lethal malignancies of the hindbrain in infants and toddlers. Lacking highly recurrent somatic mutations, PFA ependymomas are proposed to be epigenetically driven tumors for which model systems are lacking. Here we demonstrate that PFA ependymomas are maintained under hypoxia, associated with restricted availability of specific metabolites to diminish histone methylation, and increase histone demethylation and acetylation at histone 3 lysine 27 (H3K27). PFA ependymomas initiate from a cell lineage in the first trimester of human development that resides in restricted oxygen. Unlike other ependymomas, transient exposure of PFA cells to ambient oxygen induces irreversible cellular toxicity. PFA tumors exhibit a low basal level of H3K27me3, and, paradoxically, inhibition of H3K27 methylation specifically disrupts PFA tumor growth. Targeting metabolism and/or the epigenome presents a unique opportunity for rational therapy for infants with PFA ependymoma. Hypoxia reprograms the cellular metabolome and epigenome to promote growth of the most lethal ependymomas.
- Published
- 2020
13. In vivo CRISPR screens identify a dual function of MEN1 in regulating tumor-microenvironment interactions.
- Author
-
Su P, Liu Y, Chen T, Xue Y, Zeng Y, Zhu G, Chen S, Teng M, Ci X, Guo M, He MY, Hao J, Chu V, Xu W, Wang S, Mehdipour P, Xu X, Marhon SA, Soares F, Pham NA, Wu BX, Her PH, Feng S, Alshamlan N, Khalil M, Krishnan R, Yu F, Chen C, Burrows F, Hakem R, Lupien M, Harding S, Lok BH, O'Brien C, Berlin A, De Carvalho DD, Brooks DG, Schramek D, Tsao MS, and He HH
- Subjects
- Animals, Female, Humans, Mice, Cell Line, Tumor, Cell Proliferation genetics, Gene Expression Regulation, Neoplastic, Myeloid-Lymphoid Leukemia Protein genetics, Neoplasms genetics, Neoplasms pathology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, CRISPR-Cas Systems, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Tumor Microenvironment
- Abstract
Functional genomic screens in two-dimensional cell culture models are limited in identifying therapeutic targets that influence the tumor microenvironment. By comparing targeted CRISPR-Cas9 screens in a two-dimensional culture with xenografts derived from the same cell line, we identified MEN1 as the top hit that confers differential dropout effects in vitro and in vivo. MEN1 knockout in multiple solid cancer types does not impact cell proliferation in vitro but significantly promotes or inhibits tumor growth in immunodeficient or immunocompetent mice, respectively. Mechanistically, MEN1 knockout redistributes MLL1 chromatin occupancy, increasing H3K4me3 at repetitive genomic regions, activating double-stranded RNA expression and increasing neutrophil and CD8
+ T cell infiltration in immunodeficient and immunocompetent mice, respectively. Pharmacological inhibition of the menin-MLL interaction reduces tumor growth in a CD8+ T cell-dependent manner. These findings reveal tumor microenvironment-dependent oncogenic and tumor-suppressive functions of MEN1 and provide a rationale for targeting MEN1 in solid cancers., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
14. Fitness Screens Map State-Specific Glioblastoma Stem Cell Vulnerabilities.
- Author
-
MacLeod G, Molaei F, Haider S, Almeida MP, Lin S, Kushida M, Sureshkumar H, Bhatti JK, Lu JQ, Schramek D, Dirks PB, and Angers S
- Abstract
Glioblastoma (GBM) is the most common and lethal primary brain tumor in adults and is driven by self-renewing glioblastoma stem cells (GSCs) that persist after therapy and seed treatment refractory recurrent tumors. GBM tumors display a high degree of intra- and inter-tumoral heterogeneity that is a prominent barrier to targeted treatment strategies. This heterogeneity extends to GSCs that exist on a gradient between two transcriptional states or subtypes termed developmental and injury-response. Drug targets for each subtype are needed to effectively target GBM. To identify conserved and subtype-specific genetic dependencies across a large and heterogeneous panel of GSCs, we designed the GBM5K targeted gRNA library and performed fitness screens in a total of 30 patient-derived GSC cultures. The focused CRISPR screens identified the most conserved subtype-specific vulnerabilities in GSCs and elucidated the functional dependency gradient existing between the developmental and injury-response states. Developmental-specific fitness genes were enriched for transcriptional regulators of neurodevelopment, whereas injury-response-specific fitness genes were highlighted by several genes implicated in integrin and focal adhesion signaling. These context-specific vulnerabilities conferred differential sensitivity to inhibitors of β1 integrin, FAK, MEK and OLIG2. Interestingly, the screens revealed that the subtype-specific signaling pathways drive differential cyclin D (CCND1 vs. CCND2) dependencies between subtypes. These data provide biological insight and mechanistic understanding of GBM heterogeneity and point to opportunities for precision targeting of defined GBM and GSC subtypes to tackle heterogeneity.
- Published
- 2024
- Full Text
- View/download PDF
15. Early rhombic lip Protogenin +ve stem cells in a human-specific neurovascular niche initiate and maintain group 3 medulloblastoma.
- Author
-
Visvanathan A, Saulnier O, Chen C, Haldipur P, Orisme W, Delaidelli A, Shin S, Millman J, Bryant A, Abeysundara N, Wu X, Hendrikse LD, Patil V, Bashardanesh Z, Golser J, Livingston BG, Nakashima T, Funakoshi Y, Ong W, Rasnitsyn A, Aldinger KA, Richman CM, Van Ommeren R, Lee JJY, Ly M, Vladoiu MC, Kharas K, Balin P, Erickson AW, Fong V, Zhang J, Suárez RA, Wang H, Huang N, Pallota JG, Douglas T, Haapasalo J, Razavi F, Silvestri E, Sirbu O, Worme S, Kameda-Smith MM, Wu X, Daniels C, MichaelRaj AK, Bhaduri A, Schramek D, Suzuki H, Garzia L, Ahmed N, Kleinman CL, Stein LD, Dirks P, Dunham C, Jabado N, Rich JN, Li W, Sorensen PH, Wechsler-Reya RJ, Weiss WA, Millen KJ, Ellison DW, Dimitrov DS, and Taylor MD
- Subjects
- Humans, Animals, Mice, Rhombencephalon metabolism, Rhombencephalon embryology, Cerebellar Neoplasms metabolism, Cerebellar Neoplasms pathology, Endothelial Cells metabolism, Stem Cell Niche, Stem Cells metabolism, Coculture Techniques, Embryonic Structures, Metencephalon embryology, Medulloblastoma pathology, Medulloblastoma metabolism, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology
- Abstract
We identify a population of Protogenin-positive (PRTG
+ve ) MYChigh NESTINlow stem cells in the four-week-old human embryonic hindbrain that subsequently localizes to the ventricular zone of the rhombic lip (RLVZ ). Oncogenic transformation of early Prtg+ve rhombic lip stem cells initiates group 3 medulloblastoma (Gr3-MB)-like tumors. PRTG+ve stem cells grow adjacent to a human-specific interposed vascular plexus in the RLVZ , a phenotype that is recapitulated in Gr3-MB but not in other types of medulloblastoma. Co-culture of Gr3-MB with endothelial cells promotes tumor stem cell growth, with the endothelial cells adopting an immature phenotype. Targeting the PRTGhigh compartment of Gr3-MB in vivo using either the diphtheria toxin system or chimeric antigen receptor T cells constitutes effective therapy. Human Gr3-MBs likely arise from early embryonic RLVZ PRTG+ve stem cells inhabiting a specific perivascular niche. Targeting the PRTGhigh compartment and/or the perivascular niche represents an approach to treat children with Gr3-MB., Competing Interests: Declaration of interests The authors declare no competing interests. A patent related to this work was submitted by University of Pittsburgh (University Docket No. 05700/F&R ref. 48881-0028P01), US 63/275,326, “Molecules that bind to Protogenin polypeptides.”, (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
16. In vivo CRISPR screens reveal SCAF1 and USP15 as drivers of pancreatic cancer.
- Author
-
Martinez S, Wu S, Geuenich M, Malik A, Weber R, Woo T, Zhang A, Jang GH, Dervovic D, Al-Zahrani KN, Tsai R, Fodil N, Gros P, Gallinger S, Neely GG, Notta F, Sendoel A, Campbell K, Elling U, and Schramek D
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Gemcitabine, Gene Expression Regulation, Neoplastic, Mutation, Ubiquitin-Specific Proteases genetics, Ubiquitin-Specific Proteases metabolism, CRISPR-Cas Systems, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology
- Abstract
Functionally characterizing the genetic alterations that drive pancreatic cancer is a prerequisite for precision medicine. Here, we perform somatic CRISPR/Cas9 mutagenesis screens to assess the transforming potential of 125 recurrently mutated pancreatic cancer genes, which revealed USP15 and SCAF1 as pancreatic tumor suppressors. Mechanistically, we find that USP15 functions in a haploinsufficient manner and that loss of USP15 or SCAF1 leads to reduced inflammatory TNFα, TGF-β and IL6 responses and increased sensitivity to PARP inhibition and Gemcitabine. Furthermore, we find that loss of SCAF1 leads to the formation of a truncated, inactive USP15 isoform at the expense of full-length USP15, functionally coupling SCAF1 and USP15. Notably, USP15 and SCAF1 alterations are observed in 31% of pancreatic cancer patients. Our results highlight the utility of in vivo CRISPR screens to integrate human cancer genomics and mouse modeling for the discovery of cancer driver genes with potential prognostic and therapeutic implications., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. Genome-wide CRISPR screens identify novel regulators of wild-type and mutant p53 stability.
- Author
-
Lü Y, Cho T, Mukherjee S, Suarez CF, Gonzalez-Foutel NS, Malik A, Martinez S, Dervovic D, Oh RH, Langille E, Al-Zahrani KN, Hoeg L, Lin ZY, Tsai R, Mbamalu G, Rotter V, Ashton-Prolla P, Moffat J, Chemes LB, Gingras AC, Oren M, Durocher D, and Schramek D
- Subjects
- Animals, Humans, Mice, Female, CRISPR-Cas Systems, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Line, Tumor, Ubiquitin Thiolesterase genetics, Ubiquitin Thiolesterase metabolism, Gene Expression Regulation, Neoplastic, Clustered Regularly Interspaced Short Palindromic Repeats, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Protein Stability, Mutation
- Abstract
Tumor suppressor p53 (TP53) is frequently mutated in cancer, often resulting not only in loss of its tumor-suppressive function but also acquisition of dominant-negative and even oncogenic gain-of-function traits. While wild-type p53 levels are tightly regulated, mutants are typically stabilized in tumors, which is crucial for their oncogenic properties. Here, we systematically profiled the factors that regulate protein stability of wild-type and mutant p53 using marker-based genome-wide CRISPR screens. Most regulators of wild-type p53 also regulate p53 mutants, except for p53 R337H regulators, which are largely private to this mutant. Mechanistically, FBXO42 emerged as a positive regulator for a subset of p53 mutants, working with CCDC6 to control USP28-mediated mutant p53 stabilization. Additionally, C16orf72/HAPSTR1 negatively regulates both wild-type p53 and all tested mutants. C16orf72/HAPSTR1 is commonly amplified in breast cancer, and its overexpression reduces p53 levels in mouse mammary epithelium leading to accelerated breast cancer. This study offers a network perspective on p53 stability regulation, potentially guiding strategies to reinforce wild-type p53 or target mutant p53 in cancer., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
18. Proteome-scale discovery of protein degradation and stabilization effectors.
- Author
-
Poirson J, Cho H, Dhillon A, Haider S, Imrit AZ, Lam MHY, Alerasool N, Lacoste J, Mizan L, Wong C, Gingras AC, Schramek D, and Taipale M
- Subjects
- Humans, Substrate Specificity, Proteolysis Targeting Chimera metabolism, Von Hippel-Lindau Tumor Suppressor Protein metabolism, Deubiquitinating Enzymes analysis, Deubiquitinating Enzymes metabolism, Protein Stability, Proteolysis, Proteome metabolism, Proteomics, Ubiquitin-Protein Ligases analysis, Ubiquitin-Protein Ligases metabolism
- Abstract
Targeted protein degradation and stabilization are promising therapeutic modalities because of their potency, versatility and their potential to expand the druggable target space
1,2 . However, only a few of the hundreds of E3 ligases and deubiquitinases in the human proteome have been harnessed for this purpose, which substantially limits the potential of the approach. Moreover, there may be other protein classes that could be exploited for protein stabilization or degradation3-5 , but there are currently no methods that can identify such effector proteins in a scalable and unbiased manner. Here we established a synthetic proteome-scale platform to functionally identify human proteins that can promote the degradation or stabilization of a target protein in a proximity-dependent manner. Our results reveal that the human proteome contains a large cache of effectors of protein stability. The approach further enabled us to comprehensively compare the activities of human E3 ligases and deubiquitinases, identify and characterize non-canonical protein degraders and stabilizers and establish that effectors have vastly different activities against diverse targets. Notably, the top degraders were more potent against multiple therapeutically relevant targets than the currently used E3 ligases cereblon and VHL. Our study provides a functional catalogue of stability effectors for targeted protein degradation and stabilization and highlights the potential of induced proximity screens for the discovery of new proximity-dependent protein modulators., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
19. Differential DNA damage repair and PARP inhibitor vulnerability of the mammary epithelial lineages.
- Author
-
Kim H, Aliar K, Tharmapalan P, McCloskey CW, Kuttanamkuzhi A, Grünwald BT, Palomero L, Mahendralingam MJ, Waas M, Mer AS, Elliott MJ, Zhang B, Al-Zahrani KN, Langille ER, Parsons M, Narala S, Hofer S, Waterhouse PD, Hakem R, Haibe-Kains B, Kislinger T, Schramek D, Cescon DW, Pujana MA, Berman HK, and Khokha R
- Published
- 2023
- Full Text
- View/download PDF
20. Mutational processes of tobacco smoking and APOBEC activity generate protein-truncating mutations in cancer genomes.
- Author
-
Adler N, Bahcheli AT, Cheng KCL, Al-Zahrani KN, Slobodyanyuk M, Pellegrina D, Schramek D, and Reimand J
- Subjects
- Humans, Mutation, Cytidine Deaminase genetics, APOBEC Deaminases genetics, APOBEC Deaminases metabolism, Tobacco Smoking, Neoplasms genetics, Neoplasms pathology
- Abstract
Mutational signatures represent a genomic footprint of endogenous and exogenous mutational processes through tumor evolution. However, their functional impact on the proteome remains incompletely understood. We analyzed the protein-coding impact of single-base substitution (SBS) signatures in 12,341 cancer genomes from 18 cancer types. Stop-gain mutations (SGMs) (i.e., nonsense mutations) were strongly enriched in SBS signatures of tobacco smoking, APOBEC cytidine deaminases, and reactive oxygen species. These mutational processes alter specific trinucleotide contexts and thereby substitute serines and glutamic acids with stop codons. SGMs frequently affect cancer hallmark pathways and tumor suppressors such as TP53 , FAT1 , and APC . Tobacco-driven SGMs in lung cancer correlate with smoking history and highlight a preventable determinant of these harmful mutations. APOBEC-driven SGMs are enriched in YTCA motifs and associate with APOBEC3A expression. Our study exposes SGM expansion as a genetic mechanism by which endogenous and carcinogenic mutational processes directly contribute to protein loss of function, oncogenesis, and tumor heterogeneity.
- Published
- 2023
- Full Text
- View/download PDF
21. A designer peptide against the EAG2-Kvβ2 potassium channel targets the interaction of cancer cells and neurons to treat glioblastoma.
- Author
-
Dong W, Fekete A, Chen X, Liu H, Beilhartz GL, Chen X, Bahrampour S, Xiong Y, Yang Q, Zhao H, Kong T, Morioka MS, Jung G, Kim JE, Schramek D, Dirks PB, Song Y, Kim TH, He Y, Wanggou S, Li X, Melnyk RA, Wang LY, and Huang X
- Subjects
- Humans, Mice, Animals, Temozolomide pharmacology, Temozolomide therapeutic use, Ether-A-Go-Go Potassium Channels therapeutic use, Disease Models, Animal, Peptides therapeutic use, Neurons pathology, Glioblastoma drug therapy
- Abstract
Glioblastoma (GBM) is an incurable brain cancer that lacks effective therapies. Here we show that EAG2 and Kvβ2, which are predominantly expressed by GBM cells at the tumor-brain interface, physically interact to form a potassium channel complex due to a GBM-enriched Kvβ2 isoform. In GBM cells, EAG2 localizes at neuron-contacting regions in a Kvβ2-dependent manner. Genetic knockdown of the EAG2-Kvβ2 complex decreases calcium transients of GBM cells, suppresses tumor growth and invasion and extends the survival of tumor-bearing mice. We engineered a designer peptide to disrupt EAG2-Kvβ2 interaction, thereby mitigating tumor growth in patient-derived xenograft and syngeneic mouse models across GBM subtypes without overt toxicity. Neurons upregulate chemoresistant genes in GBM cells in an EAG2-Kvβ2-dependent manner. The designer peptide targets neuron-associated GBM cells and possesses robust efficacy in treating temozolomide-resistant GBM. Our findings may lead to the next-generation therapeutic agent to benefit patients with GBM., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
22. Developmental priming of cancer susceptibility.
- Author
-
Panzeri I, Fagnocchi L, Apostle S, Tompkins M, Wolfrum E, Madaj Z, Hostetter G, Liu Y, Schaefer K, Chih-Hsiang Y, Bergsma A, Drougard A, Dror E, Chandler D, Schramek D, Triche TJ Jr, and Pospisilik JA
- Abstract
DNA mutations are necessary drivers of cancer, yet only a small subset of mutated cells go on to cause the disease. To date, the mechanisms that determine which rare subset of cells transform and initiate tumorigenesis remain unclear. Here, we take advantage of a unique model of intrinsic developmental heterogeneity ( Trim28
+/ D9 ) and demonstrate that stochastic early life epigenetic variation can trigger distinct cancer-susceptibility 'states' in adulthood. We show that these developmentally primed states are characterized by differential methylation patterns at typically silenced heterochromatin, and that these epigenetic signatures are detectable as early as 10 days of age. The differentially methylated loci are enriched for genes with known oncogenic potential. These same genes are frequently mutated in human cancers, and their dysregulation correlates with poor prognosis. These results provide proof-of-concept that intrinsic developmental heterogeneity can prime individual, life-long cancer risk., Competing Interests: DECLARATION OF INTERESTS The authors declare no competing interests.- Published
- 2023
- Full Text
- View/download PDF
23. Author Correction: In vivo CRISPR screens reveal Serpinb9 and Adam2 as regulators of immune therapy response in lung cancer.
- Author
-
Dervovic D, Malik AA, Chen ELY, Narimatsu M, Adler N, Afiuni-Zadeh S, Krenbek D, Martinez S, Tsai R, Boucher J, Berman JM, Teng K, Ayyaz A, Lü Y, Mbamalu G, Loganathan SK, Lee J, Zhang L, Guidos C, Wrana J, Valipour A, Roux PP, Reimand J, Jackson HW, and Schramek D
- Published
- 2023
- Full Text
- View/download PDF
24. In vivo CRISPR screens reveal Serpinb9 and Adam2 as regulators of immune therapy response in lung cancer.
- Author
-
Dervovic D, Malik AA, Chen ELY, Narimatsu M, Adler N, Afiuni-Zadeh S, Krenbek D, Martinez S, Tsai R, Boucher J, Berman JM, Teng K, Ayyaz A, Lü Y, Mbamalu G, Loganathan SK, Lee J, Zhang L, Guidos C, Wrana J, Valipour A, Roux PP, Reimand J, Jackson HW, and Schramek D
- Subjects
- Animals, Humans, Male, Mice, Antigens, Neoplasm, Immunotherapy, Membrane Proteins genetics, T-Lymphocytes, Cytotoxic, Tumor Microenvironment, Antineoplastic Agents, Fertilins genetics, Lung Neoplasms genetics, Lung Neoplasms therapy, Serpins genetics
- Abstract
How the genetic landscape governs a tumor's response to immunotherapy remains poorly understood. To assess the immune-modulatory capabilities of 573 genes associated with altered cytotoxicity in human cancers, here we perform CRISPR/Cas9 screens directly in mouse lung cancer models. We recover the known immune evasion factors Stat1 and Serpinb9 and identify the cancer testis antigen Adam2 as an immune modulator, whose expression is induced by Kras
G12D and further elevated by immunotherapy. Using loss- and gain-of-function experiments, we show that ADAM2 functions as an oncogene by restraining interferon and TNF cytokine signaling causing reduced presentation of tumor-associated antigens. ADAM2 also restricts expression of the immune checkpoint inhibitors PDL1, LAG3, TIGIT and TIM3 in the tumor microenvironment, which might explain why ex vivo expanded and adoptively transferred cytotoxic T-cells show enhanced cytotoxic efficacy in ADAM2 overexpressing tumors. Together, direct in vivo CRISPR/Cas9 screens can uncover genetic alterations that control responses to immunotherapies., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
25. Author Correction: The stress kinase MKK7 couples oncogenic stress to p53 stability and tumor suppression.
- Author
-
Schramek D, Kotsinas A, Meixner A, Wada T, Elling U, Pospisilik JA, Neely GG, Zwick RH, Sigl V, Forni G, Serrano M, Gorgoulis VG, and Penninger JM
- Published
- 2023
- Full Text
- View/download PDF
26. In Vivo Screening Unveils Pervasive RNA-Binding Protein Dependencies in Leukemic Stem Cells and Identifies ELAVL1 as a Therapeutic Target.
- Author
-
Vujovic A, de Rooij L, Chahi AK, Chen HT, Yee BA, Loganathan SK, Liu L, Chan DCH, Tajik A, Tsao E, Moreira S, Joshi P, Xu J, Wong N, Balde Z, Jahangiri S, Zandi S, Aigner S, Dick JE, Minden MD, Schramek D, Yeo GW, and Hope KJ
- Subjects
- Humans, Cell Differentiation, Hematopoietic Stem Cells metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, RNA-Binding Proteins therapeutic use, Mitochondrial Precursor Protein Import Complex Proteins, ELAV-Like Protein 1 genetics, ELAV-Like Protein 1 metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute drug therapy
- Abstract
Acute myeloid leukemia (AML) is fueled by leukemic stem cells (LSC) whose determinants are challenging to discern from hematopoietic stem cells (HSC) or uncover by approaches focused on general cell properties. We have identified a set of RNA-binding proteins (RBP) selectively enriched in human AML LSCs. Using an in vivo two-step CRISPR-Cas9 screen to assay stem cell functionality, we found 32 RBPs essential for LSCs in MLL-AF9;NrasG12D AML. Loss-of-function approaches targeting key hit RBP ELAVL1 compromised LSC-driven in vivo leukemic reconstitution, and selectively depleted primitive malignant versus healthy cells. Integrative multiomics revealed differentiation, splicing, and mitochondrial metabolism as key features defining the leukemic ELAVL1-mRNA interactome with mitochondrial import protein, TOMM34, being a direct ELAVL1-stabilized target whose repression impairs AML propagation. Altogether, using a stem cell-adapted in vivo CRISPR screen, this work demonstrates pervasive reliance on RBPs as regulators of LSCs and highlights their potential as therapeutic targets in AML., Significance: LSC-targeted therapies remain a significant unmet need in AML. We developed a stem-cell-adapted in vivo CRISPR screen to identify key LSC drivers. We uncover widespread RNA-binding protein dependencies in LSCs, including ELAVL1, which we identify as a novel therapeutic vulnerability through its regulation of mitochondrial metabolism. This article is highlighted in the In This Issue feature, p. 171., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
27. The NOTCH-RIPK4-IRF6-ELOVL4 Axis Suppresses Squamous Cell Carcinoma.
- Author
-
Yan Y, Gauthier MA, Malik A, Fotiadou I, Ostrovski M, Dervovic D, Ghadban L, Tsai R, Gish G, Loganathan SK, and Schramek D
- Abstract
Receptor-interacting serine/threonine protein kinase 4 (RIPK4) and its kinase substrate the transcription factor interferon regulatory factor 6 (IRF6) play critical roles in the development and maintenance of the epidermis. In addition, ourselves and others have previously shown that RIPK4 is a NOTCH target gene that suppresses the development of cutaneous and head and neck squamous cell carcinomas (HNSCCs). In this study, we used autochthonous mouse models, where the expression of Pik3ca
H1047R oncogene predisposes the skin and oral cavity to tumor development, and show that not only loss of Ripk4 , but also loss of its kinase substrate Irf6 , triggers rapid SCC development. In vivo rescue experiments using Ripk4 or a kinase-dead Ripk4 mutant showed that the tumor suppressive function of Ripk4 is dependent on its kinase activity. To elucidate critical mediators of this tumor suppressive pathway, we performed transcriptional profiling of Ripk4 -deficient epidermal cells followed by multiplexed in vivo CRISPR screening to identify genes with tumor suppressive capabilities. We show that Elovl4 is a critical Notch-Ripk4-Irf6 downstream target gene, and that Elovl4 loss itself triggers SCC development. Importantly, overexpression of Elovl4 suppressed tumor growth of Ripk4 -deficient keratinocytes. Altogether, our work identifies a potent Notch1-Ripk4-Irf6-Elovl4 tumor suppressor axis.- Published
- 2023
- Full Text
- View/download PDF
28. Loss of Epigenetic Regulation Disrupts Lineage Integrity, Induces Aberrant Alveogenesis, and Promotes Breast Cancer.
- Author
-
Langille E, Al-Zahrani KN, Ma Z, Liang M, Uuskula-Reimand L, Espin R, Teng K, Malik A, Bergholtz H, Ghamrasni SE, Afiuni-Zadeh S, Tsai R, Alvi S, Elia A, Lü Y, Oh RH, Kozma KJ, Trcka D, Narimatsu M, Liu JC, Nguyen T, Barutcu S, Loganathan SK, Bremner R, Bader GD, Egan SE, Cescon DW, Sørlie T, Wrana JL, Jackson HW, Wilson MD, Witkiewicz AK, Knudsen ES, Pujana MA, Wahl GM, and Schramek D
- Subjects
- Humans, Mice, Animals, Female, Epigenesis, Genetic, Neoplasm Recurrence, Local genetics, Cell Transformation, Neoplastic genetics, Breast Neoplasms pathology, Carcinoma, Intraductal, Noninfiltrating genetics
- Abstract
Systematically investigating the scores of genes mutated in cancer and discerning disease drivers from inconsequential bystanders is a prerequisite for precision medicine but remains challenging. Here, we developed a somatic CRISPR/Cas9 mutagenesis screen to study 215 recurrent "long-tail" breast cancer genes, which revealed epigenetic regulation as a major tumor-suppressive mechanism. We report that components of the BAP1 and COMPASS-like complexes, including KMT2C/D, KDM6A, BAP1, and ASXL1/2 ("EpiDrivers"), cooperate with PIK3CAH1047R to transform mouse and human breast epithelial cells. Mechanistically, we find that activation of PIK3CAH1047R and concomitant EpiDriver loss triggered an alveolar-like lineage conversion of basal mammary epithelial cells and accelerated formation of luminal-like tumors, suggesting a basal origin for luminal tumors. EpiDriver mutations are found in ∼39% of human breast cancers, and ∼50% of ductal carcinoma in situ express casein, suggesting that lineage infidelity and alveogenic mimicry may significantly contribute to early steps of breast cancer etiology., Significance: Infrequently mutated genes comprise most of the mutational burden in breast tumors but are poorly understood. In vivo CRISPR screening identified functional tumor suppressors that converged on epigenetic regulation. Loss of epigenetic regulators accelerated tumorigenesis and revealed lineage infidelity and aberrant expression of alveogenesis genes as potential early events in tumorigenesis. This article is highlighted in the In This Issue feature, p. 2711., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
29. A CRISPR Path to Finding Vulnerabilities and Solving Drug Resistance: Targeting the Diverse Cancer Landscape and Its Ecosystem.
- Author
-
McLean B, Istadi A, Clack T, Vankan M, Schramek D, Neely GG, and Pajic M
- Abstract
Cancer is the second leading cause of death globally, with therapeutic resistance being a major cause of treatment failure in the clinic. The dynamic signaling that occurs between tumor cells and the diverse cells of the surrounding tumor microenvironment actively promotes disease progression and therapeutic resistance. Improving the understanding of how tumors evolve following therapy and the molecular mechanisms underpinning de novo or acquired resistance is thus critical for the identification of new targets and for the subsequent development of more effective combination regimens. Simultaneously targeting multiple hallmark capabilities of cancer to circumvent adaptive or evasive resistance may lead to significantly improved treatment response in the clinic. Here, the latest applications of functional genomics tools, such as clustered regularly interspaced short palindromic repeats (CRISPR) editing, to characterize the dynamic cancer resistance mechanisms, from improving the understanding of resistance to classical chemotherapeutics, to deciphering unique mechanisms that regulate tumor responses to new targeted agents and immunotherapies, are discussed. Potential avenues of future research in combating therapeutic resistance, the contribution of tumor-stroma signaling in this setting, and how advanced functional genomics tools can help streamline the identification of key molecular determinants of drug response are explored., Competing Interests: The authors declare no conflict of interest., (© 2022 The Authors. Advanced Genetics published by Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
30. A noncoding single-nucleotide polymorphism at 8q24 drives IDH1 -mutant glioma formation.
- Author
-
Yanchus C, Drucker KL, Kollmeyer TM, Tsai R, Winick-Ng W, Liang M, Malik A, Pawling J, De Lorenzo SB, Ali A, Decker PA, Kosel ML, Panda A, Al-Zahrani KN, Jiang L, Browning JWL, Lowden C, Geuenich M, Hernandez JJ, Gosio JT, Ahmed M, Loganathan SK, Berman J, Trcka D, Michealraj KA, Fortin J, Carson B, Hollingsworth EW, Jacinto S, Mazrooei P, Zhou L, Elia A, Lupien M, He HH, Murphy DJ, Wang L, Abyzov A, Dennis JW, Maass PG, Campbell K, Wilson MD, Lachance DH, Wrensch M, Wiencke J, Mak T, Pennacchio LA, Dickel DE, Visel A, Wrana J, Taylor MD, Zadeh G, Dirks P, Eckel-Passow JE, Attisano L, Pombo A, Ida CM, Kvon EZ, Jenkins RB, and Schramek D
- Subjects
- Animals, Humans, Mice, Mutation, Polymorphism, Single Nucleotide, Brain Neoplasms genetics, Brain Neoplasms pathology, Chromosomes, Human, Pair 8 genetics, Glioma genetics, Glioma pathology, Isocitrate Dehydrogenase genetics
- Abstract
Establishing causal links between inherited polymorphisms and cancer risk is challenging. Here, we focus on the single-nucleotide polymorphism rs55705857, which confers a sixfold greater risk of isocitrate dehydrogenase ( IDH) -mutant low-grade glioma (LGG). We reveal that rs55705857 itself is the causal variant and is associated with molecular pathways that drive LGG. Mechanistically, we show that rs55705857 resides within a brain-specific enhancer, where the risk allele disrupts OCT2/4 binding, allowing increased interaction with the Myc promoter and increased Myc expression. Mutating the orthologous mouse rs55705857 locus accelerated tumor development in an Idh1
R132H -driven LGG mouse model from 472 to 172 days and increased penetrance from 30% to 75%. Our work reveals mechanisms of the heritable predisposition to lethal glioma in ~40% of LGG patients.- Published
- 2022
- Full Text
- View/download PDF
31. Cross-talk between mutant p53 and p62/SQSTM1 augments cancer cell migration by promoting the degradation of cell adhesion proteins.
- Author
-
Mukherjee S, Maddalena M, Lü Y, Martinez S, Nataraj NB, Noronha A, Sinha S, Teng K, Cohen-Kaplan V, Ziv T, Arandkar S, Hassin O, Chatterjee R, Pirona AC, Shreberk-Shaked M, Gershoni A, Aylon Y, Elazar Z, Yarden Y, Schramek D, and Oren M
- Subjects
- Cell Adhesion genetics, Cell Line, Tumor, Genes, p53, Humans, Mutation, Sequestosome-1 Protein genetics, Sequestosome-1 Protein metabolism, Cell Movement genetics, Pancreatic Neoplasms genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Missense mutations in the p53 tumor suppressor abound in human cancer. Common (“hotspot”) mutations endow mutant p53 (mutp53) proteins with oncogenic gain of function (GOF), including enhanced cell migration and invasiveness, favoring cancer progression. GOF is usually attributed to transcriptional effects of mutp53. To elucidate transcription-independent effects of mutp53, we characterized the protein interactome of the p53R273H mutant in cells derived from pancreatic ductal adenocarcinoma (PDAC), where p53R273H is the most frequent p53 mutant. We now report that p53R273H, but not the p53R175H hotspot mutant, interacts with SQSTM1/p62 and promotes cancer cell migration and invasion in a p62-dependent manner. Mechanistically, the p53R273H-p62 axis drives the proteasomal degradation of several cell junction–associated proteins, including the gap junction protein Connexin 43, facilitating scattered cell migration. Concordantly, down-regulation of Connexin 43 augments PDAC cell migration, while its forced overexpression blunts the promigratory effect of the p53R273H-p62 axis. These findings define a mechanism of mutp53 GOF.
- Published
- 2022
- Full Text
- View/download PDF
32. CDK/cyclin dependencies define extreme cancer cell-cycle heterogeneity and collateral vulnerabilities.
- Author
-
Knudsen ES, Kumarasamy V, Nambiar R, Pearson JD, Vail P, Rosenheck H, Wang J, Eng K, Bremner R, Schramek D, Rubin SM, Welm AL, and Witkiewicz AK
- Subjects
- Cell Cycle genetics, Cell Cycle Proteins genetics, Cell Division, Cyclin-Dependent Kinase 4, Cyclin-Dependent Kinase Inhibitor p27 genetics, Humans, Cyclin-Dependent Kinases metabolism, Neoplasms genetics
- Abstract
Progression through G1/S phase of the cell cycle is coordinated by cyclin-dependent kinase (CDK) activities. Here, we find that the requirement for different CDK activities and cyclins in driving cancer cell cycles is highly heterogeneous. The differential gene requirements associate with tumor origin and genetic alterations. We define multiple mechanisms for G1/S progression in RB-proficient models, which are CDK4/6 independent and elicit resistance to FDA-approved inhibitors. Conversely, RB-deficient models are intrinsically CDK4/6 independent, but exhibit differential requirements for cyclin E. These dependencies for CDK and cyclins associate with gene expression programs that denote intrinsically different cell-cycle states. Mining therapeutic sensitivities shows that there are reciprocal vulnerabilities associated with RB1 or CCND1 expression versus CCNE1 or CDKN2A. Together, these findings illustrate the complex nature of cancer cell cycles and the relevance for precision therapeutic intervention., Competing Interests: Declaration of interests A.L.W. has received royalties from licenses of patient-derived xenograft or organoid models issued by the University of Utah. The university may issue new licenses in the future at its discretion, which may result in additional royalties. E.S.K. and A.K.W. have served on the BioVica scientific advisory board., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
33. Pan-cancer analysis of non-coding transcripts reveals the prognostic onco-lncRNA HOXA10-AS in gliomas.
- Author
-
Isaev K, Jiang L, Wu S, Lee CA, Watters V, Fort V, Tsai R, Coutinho FJ, Hussein SMI, Zhang J, Wu J, Dirks PB, Schramek D, and Reimand J
- Subjects
- Animals, Biomarkers, Tumor genetics, Brain Neoplasms genetics, Brain Neoplasms pathology, Cell Line, Tumor, Cell Movement, Cell Proliferation, Databases, Genetic, Gene Expression Regulation, Neoplastic, Glioma genetics, Glioma pathology, Humans, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase metabolism, Machine Learning, Mice, Inbred NOD, Mice, SCID, Mutation, Neoplasm Invasiveness, Predictive Value of Tests, Prognosis, RNA, Long Noncoding genetics, Reproducibility of Results, Signal Transduction, Mice, Biomarkers, Tumor metabolism, Brain Neoplasms metabolism, Gene Expression Profiling, Glioma metabolism, RNA, Long Noncoding metabolism, Transcriptome
- Abstract
Long non-coding RNAs (lncRNAs) are increasingly recognized as functional units in cancer and powerful biomarkers; however, most remain uncharacterized. Here, we analyze 5,592 prognostic lncRNAs in 9,446 cancers of 30 types using machine learning. We identify 166 lncRNAs whose expression correlates with survival and improves the accuracy of common clinical variables, molecular features, and cancer subtypes. Prognostic lncRNAs are often characterized by switch-like expression patterns. In low-grade gliomas, HOXA10-AS activation is a robust marker of poor prognosis that complements IDH1/2 mutations, as validated in another retrospective cohort, and correlates with developmental pathways in tumor transcriptomes. Loss- and gain-of-function studies in patient-derived glioma cells, organoids, and xenograft models identify HOXA10-AS as a potent onco-lncRNA that regulates cell proliferation, contact inhibition, invasion, Hippo signaling, and mitotic and neuro-developmental pathways. Our study underscores the pan-cancer potential of the non-coding transcriptome for identifying biomarkers and regulators of cancer progression., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
34. Binary pan-cancer classes with distinct vulnerabilities defined by pro- or anti-cancer YAP/TEAD activity.
- Author
-
Pearson JD, Huang K, Pacal M, McCurdy SR, Lu S, Aubry A, Yu T, Wadosky KM, Zhang L, Wang T, Gregorieff A, Ahmad M, Dimaras H, Langille E, Cole SPC, Monnier PP, Lok BH, Tsao MS, Akeno N, Schramek D, Wikenheiser-Brokamp KA, Knudsen ES, Witkiewicz AK, Wrana JL, Goodrich DW, and Bremner R
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Enhancer Elements, Genetic, Gene Expression Regulation, Neoplastic, Humans, Integrins metabolism, Male, Mice, Transgenic, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Retinal Neoplasms genetics, Retinal Neoplasms pathology, Retinoblastoma genetics, Retinoblastoma pathology, Retinoblastoma Binding Proteins genetics, TEA Domain Transcription Factors metabolism, Ubiquitin-Protein Ligases genetics, Xenograft Model Antitumor Assays, Lung Neoplasms genetics, Small Cell Lung Carcinoma genetics, TEA Domain Transcription Factors genetics, Transcriptional Coactivator with PDZ-Binding Motif Proteins genetics, YAP-Signaling Proteins genetics
- Abstract
Cancer heterogeneity impacts therapeutic response, driving efforts to discover over-arching rules that supersede variability. Here, we define pan-cancer binary classes based on distinct expression of YAP and YAP-responsive adhesion regulators. Combining informatics with in vivo and in vitro gain- and loss-of-function studies across multiple murine and human tumor types, we show that opposite pro- or anti-cancer YAP activity functionally defines binary YAP
on or YAPoff cancer classes that express or silence YAP, respectively. YAPoff solid cancers are neural/neuroendocrine and frequently RB1-/- , such as retinoblastoma, small cell lung cancer, and neuroendocrine prostate cancer. YAP silencing is intrinsic to the cell of origin, or acquired with lineage switching and drug resistance. The binary cancer groups exhibit distinct YAP-dependent adhesive behavior and pharmaceutical vulnerabilities, underscoring clinical relevance. Mechanistically, distinct YAP/TEAD enhancers in YAPoff or YAPon cancers deploy anti-cancer integrin or pro-cancer proliferative programs, respectively. YAP is thus pivotal across cancer, but in opposite ways, with therapeutic implications., Competing Interests: Declaration of interests B.H.L. reports honoraria and non-financial support from AstraZeneca, and has received research funding from Pfizer and AstraZeneca not related to this work., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
35. AJUBA: A regulator of epidermal homeostasis and cancer.
- Author
-
Schleicher K and Schramek D
- Subjects
- Animals, Cell Differentiation physiology, Cell Proliferation physiology, Humans, Signal Transduction physiology, Epidermis metabolism, Homeostasis physiology, LIM Domain Proteins metabolism, Neoplasms physiopathology, Regeneration physiology
- Abstract
The epidermis, outermost layer of the skin, is constantly renewing itself through proliferative and differentiation processes. These processes are vital to maintain proper epidermal integrity during skin development and homeostasis and for preventing skin diseases and cancers. The biological mechanisms that permit this balancing act are vast, where individual pathway regulators are known, but the exact regulatory control and cross-talk between simultaneously turning one biological pathway on and an opposing one off remain elusive. This review explores the diverse roles the scaffolding protein AJUBA plays during epidermal homeostasis and cancer. Initially identified for its role in promoting meiotic progression in oocytes through Grb2 and MAP kinase activity, AJUBA also maintains cytoskeletal tension permitting epidermal tissue development and responds to retinoic acid committing cells to initiate development of surface epidermal layer. AJUBA regulates proliferation of skin stem cells through Hippo and Wnt signalling and encourages mitotic commitment through Aurora-A, Aurora-B and CDK1. In addition, AJUBA also induces epidermal differentiation to maintain appropriate epidermal thickness and barrier function by activating Notch signalling and stabilizing catenins and actin during cellular remodelling. AJUBA also plays an imperative context-dependent tumor-promoting and tumor-suppressive role within epithelial cancers. AJUBA's abundant roles within the epidermis signify its importance as a molecular switchboard, vetting multiple signalling pathways to control epidermal biology., (© 2020 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
36. Prevalence of the Brazilian TP53 Founder c.1010G>A (p.Arg337His) in Lung Adenocarcinoma: Is Genotyping Warranted in All Brazilian Patients?
- Author
-
Vieira IA, Andreis TF, Fernandes BV, Achatz MI, Macedo GS, Schramek D, and Ashton-Prolla P
- Abstract
In Southern and Southeastern Brazil, there is a germline pathogenic variant with incomplete penetrance located in the oligomerization domain of TP53 , c.1010G>A (p.Arg337His). Due to a founder effect, the variant is present in 0.3% of the general population of the region. Recently, this variant was identified in 4.4 and 8.9% of two apparently unselected, single center case series of Brazilian lung adenocarcinoma (LUAD) patients from the Southeastern and Central regions of the country, respectively. In the present study, our aim was to examine TP53 c.1010G>A allele and genotype frequencies in LUAD samples obtained from patients diagnosed in Southern Brazil. A total of 586 LUAD samples (tumor DNA) recruited from multiple centers in the region were tested, and the mutant allele was identified using TaqMan
® assays in seven cases (7/586, 1.2%) which were submitted to next generation sequencing analyses for confirmation. Somatic EGFR mutations were more frequent in TP53 c.1010G>A carriers than in non-carriers (57.1 vs. 17.6%, respectively). Further studies are needed to confirm if TP53 c.1010G>A is a driver in LUAD carcinogenesis and to verify if there is a combined effect of EGFR and germline TP53 c.1010G>A. Although variant frequency was higher than observed in the general population, it is less than previously reported in LUAD patients from other Brazilian regions. Additional data, producing regional allele frequency information in larger series of patients and including cost-effectiveness analyses, are necessary to determine if TP53 c.1010G>A screening in all Brazilian LUAD patients is justified., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Vieira, Andreis, Fernandes, Achatz, Macedo, Schramek and Ashton-Prolla.)- Published
- 2021
- Full Text
- View/download PDF
37. An Adaptive Physiologically Based Pharmacokinetic-Driven Design to Investigate the Effect of Itraconazole and Rifampicin on the Pharmacokinetics of Molibresib (GSK525762) in Healthy Female Volunteers.
- Author
-
Riddell K, Patel A, Collins G, Zhou Y, Schramek D, Kremer BE, and Ferron-Brady G
- Subjects
- Adult, Area Under Curve, Cross-Over Studies, Dose-Response Relationship, Drug, Drug Interactions, Female, Healthy Volunteers, Humans, Metabolic Clearance Rate, Middle Aged, Benzodiazepines pharmacokinetics, Cytochrome P-450 CYP3A Inducers pharmacology, Cytochrome P-450 CYP3A Inhibitors pharmacology, Itraconazole pharmacology, Rifampin pharmacology
- Abstract
Molibresib (GSK525762), an orally bioavailable small molecule with 2 major equipotent active metabolites, is being developed for the treatment of cancers. Molibresib is a substrate of cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp). To enable administering safe doses of molibresib to healthy volunteers, this 2-part randomized, open-label, crossover drug-drug interaction trial was conducted as an adaptive design study using physiologically based pharmacokinetic (PBPK) modeling and simulation to predict the lowest doses of molibresib that could be safely administered alone (10 mg) or with itraconazole and rifampicin (strong inhibitors and inducers of CYP3A and P-gp, respectively). PBPK simulation guided the molibresib dose (5 mg) to be administered along with itraconazole in part 1. Itraconazole increased total exposure (AUC) of molibresib by 4.15-fold with a 66% increase in C
max , whereas the total AUC and Cmax for the 2 major active metabolites of molibresib decreased by about 70% and 87%, respectively. A second PBPK simulation was conducted with part 1 data to also include the active metabolites to update the recommendation for the molibresib dose (20 mg) with rifampicin. With rifampicin, the AUC and Cmax of molibresib decreased by approximately 91% and 80%, respectively, whereas the AUC of the 2 active metabolites decreased to a lesser extent (8%), with a 2-fold increase in Cmax . The results of this study confirmed the in vitro data that molibresib is a substrate for CYP3A4. The adaptive design, including Simcyp simulations, allowed evaluation of 2 drug interactions of an oncology drug in a single trial, thus minimizing time and exposures administered to healthy subjects., (© 2020 Glaxo Group Limited. The Journal of Clinical Pharmacology published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.)- Published
- 2021
- Full Text
- View/download PDF
38. In Vivo CRISPR/Cas9 Screening to Simultaneously Evaluate Gene Function in Mouse Skin and Oral Cavity.
- Author
-
Loganathan SK, Malik A, Langille E, Luxenburg C, and Schramek D
- Subjects
- Animals, Base Sequence, CRISPR-Associated Protein 9 genetics, Epithelium metabolism, High-Throughput Nucleotide Sequencing, Integrases metabolism, Mice, Microinjections, Phenotype, Ultrasonics, CRISPR-Associated Protein 9 metabolism, CRISPR-Cas Systems genetics, Genetic Testing, Mouth metabolism, Skin metabolism
- Abstract
Genetically modified mouse models (GEMM) have been instrumental in assessing gene function, modeling human diseases, and serving as preclinical model to assess therapeutic avenues. However, their time-, labor- and cost-intensive nature limits their utility for systematic analysis of gene function. Recent advances in genome-editing technologies overcome those limitations and allow for the rapid generation of specific gene perturbations directly within specific mouse organs in a multiplexed and rapid manner. Here, we describe a CRISPR/Cas9-based method (Clustered Regularly Interspaced Short Palindromic Repeats) to generate thousands of gene knock-out clones within the epithelium of the skin and oral cavity of mice, and provide a protocol detailing the steps necessary to perform a direct in vivo CRISPR screen for tumor suppressor genes. This approach can be applied to other organs or other CRISPR/Cas9 technologies such as CRISPR-activation or CRISPR-inactivation to study the biological function of genes during tissue homeostasis or in various disease settings.
- Published
- 2020
- Full Text
- View/download PDF
39. The GATOR-Rag GTPase pathway inhibits mTORC1 activation by lysosome-derived amino acids.
- Author
-
Hesketh GG, Papazotos F, Pawling J, Rajendran D, Knight JDR, Martinez S, Taipale M, Schramek D, Dennis JW, and Gingras AC
- Subjects
- Enzyme Activation, HEK293 Cells, Humans, Pinocytosis, Proteolysis, Amino Acids metabolism, GTP Phosphohydrolases metabolism, Lysosomes enzymology, Mechanistic Target of Rapamycin Complex 1 metabolism, R-SNARE Proteins metabolism
- Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) couples nutrient sufficiency to cell growth. mTORC1 is activated by exogenously acquired amino acids sensed through the GATOR-Rag guanosine triphosphatase (GTPase) pathway, or by amino acids derived through lysosomal degradation of protein by a poorly defined mechanism. Here, we revealed that amino acids derived from the degradation of protein (acquired through oncogenic Ras-driven macropinocytosis) activate mTORC1 by a Rag GTPase-independent mechanism. mTORC1 stimulation through this pathway required the HOPS complex and was negatively regulated by activation of the GATOR-Rag GTPase pathway. Therefore, distinct but functionally coordinated pathways control mTORC1 activity on late endocytic organelles in response to distinct sources of amino acids., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
40. Copper bioavailability is a KRAS-specific vulnerability in colorectal cancer.
- Author
-
Aubert L, Nandagopal N, Steinhart Z, Lavoie G, Nourreddine S, Berman J, Saba-El-Leil MK, Papadopoli D, Lin S, Hart T, Macleod G, Topisirovic I, Gaboury L, Fahrni CJ, Schramek D, Meloche S, Angers S, and Roux PP
- Subjects
- Animals, Biological Availability, CRISPR-Cas Systems, Cell Line, Tumor, Cell Proliferation, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Copper-Transporting ATPases metabolism, Female, Humans, Intestinal Mucosa pathology, Mice, Mice, Knockout, Mice, Nude, Mice, SCID, Mutation, Colorectal Neoplasms metabolism, Copper metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism
- Abstract
Despite its importance in human cancers, including colorectal cancers (CRC), oncogenic KRAS has been extremely challenging to target therapeutically. To identify potential vulnerabilities in KRAS-mutated CRC, we characterize the impact of oncogenic KRAS on the cell surface of intestinal epithelial cells. Here we show that oncogenic KRAS alters the expression of a myriad of cell-surface proteins implicated in diverse biological functions, and identify many potential surface-accessible therapeutic targets. Cell surface-based loss-of-function screens reveal that ATP7A, a copper-exporter upregulated by mutant KRAS, is essential for neoplastic growth. ATP7A is upregulated at the surface of KRAS-mutated CRC, and protects cells from excess copper-ion toxicity. We find that KRAS-mutated cells acquire copper via a non-canonical mechanism involving macropinocytosis, which appears to be required to support their growth. Together, these results indicate that copper bioavailability is a KRAS-selective vulnerability that could be exploited for the treatment of KRAS-mutated neoplasms.
- Published
- 2020
- Full Text
- View/download PDF
41. Recognition of Semaphorin Proteins by P. sordellii Lethal Toxin Reveals Principles of Receptor Specificity in Clostridial Toxins.
- Author
-
Lee H, Beilhartz GL, Kucharska I, Raman S, Cui H, Lam MHY, Liang H, Rubinstein JL, Schramek D, Julien JP, Melnyk RA, and Taipale M
- Subjects
- Animals, Bacterial Toxins chemistry, Bacterial Toxins toxicity, Binding Sites, CRISPR-Cas Systems genetics, Cell Line, Cryoelectron Microscopy, Edema pathology, Edema prevention & control, Female, Humans, Lung drug effects, Lung pathology, Mice, Mice, Inbred C57BL, Molecular Dynamics Simulation, Mutagenesis, Site-Directed, Protein Binding, Protein Structure, Tertiary, Recombinant Proteins biosynthesis, Recombinant Proteins isolation & purification, Recombinant Proteins therapeutic use, Semaphorins chemistry, Semaphorins genetics, Bacterial Toxins metabolism, Clostridium sordellii metabolism, Semaphorins metabolism
- Abstract
Pathogenic clostridial species secrete potent toxins that induce severe host tissue damage. Paeniclostridium sordellii lethal toxin (TcsL) causes an almost invariably lethal toxic shock syndrome associated with gynecological infections. TcsL is 87% similar to C. difficile TcdB, which enters host cells via Frizzled receptors in colon epithelium. However, P. sordellii infections target vascular endothelium, suggesting that TcsL exploits another receptor. Here, using CRISPR/Cas9 screening, we establish semaphorins SEMA6A and SEMA6B as TcsL receptors. We demonstrate that recombinant SEMA6A can protect mice from TcsL-induced edema. A 3.3 Å cryo-EM structure shows that TcsL binds SEMA6A with the same region that in TcdB binds structurally unrelated Frizzled. Remarkably, 15 mutations in this evolutionarily divergent surface are sufficient to switch binding specificity of TcsL to that of TcdB. Our findings establish semaphorins as physiologically relevant receptors for TcsL and reveal the molecular basis for the difference in tissue targeting and disease pathogenesis between highly related toxins., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
42. Metabolic Regulation of the Epigenome Drives Lethal Infantile Ependymoma.
- Author
-
Michealraj KA, Kumar SA, Kim LJY, Cavalli FMG, Przelicki D, Wojcik JB, Delaidelli A, Bajic A, Saulnier O, MacLeod G, Vellanki RN, Vladoiu MC, Guilhamon P, Ong W, Lee JJY, Jiang Y, Holgado BL, Rasnitsyn A, Malik AA, Tsai R, Richman CM, Juraschka K, Haapasalo J, Wang EY, De Antonellis P, Suzuki H, Farooq H, Balin P, Kharas K, Van Ommeren R, Sirbu O, Rastan A, Krumholtz SL, Ly M, Ahmadi M, Deblois G, Srikanthan D, Luu B, Loukides J, Wu X, Garzia L, Ramaswamy V, Kanshin E, Sánchez-Osuna M, El-Hamamy I, Coutinho FJ, Prinos P, Singh S, Donovan LK, Daniels C, Schramek D, Tyers M, Weiss S, Stein LD, Lupien M, Wouters BG, Garcia BA, Arrowsmith CH, Sorensen PH, Angers S, Jabado N, Dirks PB, Mack SC, Agnihotri S, Rich JN, and Taylor MD
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Line, Cell Proliferation genetics, DNA Methylation genetics, Epigenomics methods, Histones genetics, Histones metabolism, Humans, Infant, Lysine genetics, Lysine metabolism, Male, Mice, Inbred C57BL, Mutation genetics, Ependymoma genetics, Ependymoma metabolism, Epigenome genetics, Infratentorial Neoplasms genetics, Infratentorial Neoplasms metabolism
- Abstract
Posterior fossa A (PFA) ependymomas are lethal malignancies of the hindbrain in infants and toddlers. Lacking highly recurrent somatic mutations, PFA ependymomas are proposed to be epigenetically driven tumors for which model systems are lacking. Here we demonstrate that PFA ependymomas are maintained under hypoxia, associated with restricted availability of specific metabolites to diminish histone methylation, and increase histone demethylation and acetylation at histone 3 lysine 27 (H3K27). PFA ependymomas initiate from a cell lineage in the first trimester of human development that resides in restricted oxygen. Unlike other ependymomas, transient exposure of PFA cells to ambient oxygen induces irreversible cellular toxicity. PFA tumors exhibit a low basal level of H3K27me3, and, paradoxically, inhibition of H3K27 methylation specifically disrupts PFA tumor growth. Targeting metabolism and/or the epigenome presents a unique opportunity for rational therapy for infants with PFA ependymoma., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
43. In vivo CRISPR screens reveal potent driver mutations in head and neck cancers.
- Author
-
Loganathan SK and Schramek D
- Abstract
We have recently tested the transforming potential of 484 'long-tail' genes, which are recurrently albeit infrequently mutated in head and neck cancers (HNSCC). We identified 15 novel tumor suppressors and our top hits converge on regulating the NOTCH signaling pathway. Therapeutic approaches activating NOTCH signaling could be a promising strategy to treat two-thirds of human HNSCC patients., (© 2020 Taylor & Francis Group, LLC.)
- Published
- 2020
- Full Text
- View/download PDF
44. Mutant ACVR1 Arrests Glial Cell Differentiation to Drive Tumorigenesis in Pediatric Gliomas.
- Author
-
Fortin J, Tian R, Zarrabi I, Hill G, Williams E, Sanchez-Duffhues G, Thorikay M, Ramachandran P, Siddaway R, Wong JF, Wu A, Apuzzo LN, Haight J, You-Ten A, Snow BE, Wakeham A, Goldhamer DJ, Schramek D, Bullock AN, Dijke PT, Hawkins C, and Mak TW
- Subjects
- Activin Receptors, Type I antagonists & inhibitors, Activin Receptors, Type I metabolism, Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Bone Morphogenetic Proteins genetics, Bone Morphogenetic Proteins metabolism, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Cell Differentiation genetics, Cell Line, Tumor, Class I Phosphatidylinositol 3-Kinases genetics, Class I Phosphatidylinositol 3-Kinases metabolism, Female, Glioma drug therapy, Glioma genetics, Histones genetics, Histones metabolism, Humans, Lactones pharmacology, Male, Mice, Transgenic, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neuroglia metabolism, Neuroglia pathology, Oligodendroglia pathology, Receptor, Platelet-Derived Growth Factor alpha genetics, Receptor, Platelet-Derived Growth Factor alpha metabolism, SOXC Transcription Factors genetics, SOXC Transcription Factors metabolism, Activin Receptors, Type I chemistry, Activin Receptors, Type I genetics, Brain Neoplasms pathology, Glioma pathology, Mutation
- Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors for which there is currently no effective treatment. Some of these tumors combine gain-of-function mutations in ACVR1, PIK3CA, and histone H3-encoding genes. The oncogenic mechanisms of action of ACVR1 mutations are currently unknown. Using mouse models, we demonstrate that Acvr1
G328V arrests the differentiation of oligodendroglial lineage cells, and cooperates with Hist1h3bK27M and Pik3caH1047R to generate high-grade diffuse gliomas. Mechanistically, Acvr1G328V upregulates transcription factors which control differentiation and DIPG cell fitness. Furthermore, we characterize E6201 as a dual inhibitor of ACVR1 and MEK1/2, and demonstrate its efficacy toward tumor cells in vivo. Collectively, our results describe an oncogenic mechanism of action for ACVR1 mutations, and suggest therapeutic strategies for DIPGs., Competing Interests: Declarations of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
45. Rare driver mutations in head and neck squamous cell carcinomas converge on NOTCH signaling.
- Author
-
Loganathan SK, Schleicher K, Malik A, Quevedo R, Langille E, Teng K, Oh RH, Rathod B, Tsai R, Samavarchi-Tehrani P, Pugh TJ, Gingras AC, and Schramek D
- Subjects
- ADAM10 Protein genetics, Amyloid Precursor Protein Secretases genetics, Animals, CRISPR-Cas Systems, Female, Genetic Testing, HEK293 Cells, Humans, LIM Domain Proteins genetics, Male, Membrane Proteins genetics, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Mutation, Receptors, Notch genetics, Signal Transduction genetics, Genes, Tumor Suppressor, Genetic Predisposition to Disease, Head and Neck Neoplasms genetics, Squamous Cell Carcinoma of Head and Neck genetics, Tumor Suppressor Proteins genetics
- Abstract
In most human cancers, only a few genes are mutated at high frequencies; most are mutated at low frequencies. The functional consequences of these recurrent but infrequent "long tail" mutations are often unknown. We focused on 484 long tail genes in head and neck squamous cell carcinoma (HNSCC) and used in vivo CRISPR to screen for genes that, upon mutation, trigger tumor development in mice. Of the 15 tumor-suppressor genes identified, ADAM10 and AJUBA suppressed HNSCC in a haploinsufficient manner by promoting NOTCH receptor signaling. ADAM10 and AJUBA mutations or monoallelic loss occur in 28% of human HNSCC cases and are mutually exclusive with NOTCH receptor mutations. Our results show that oncogenic mutations in 67% of human HNSCC cases converge onto the NOTCH signaling pathway, making NOTCH inactivation a hallmark of HNSCC., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
46. CRISPR-Switch regulates sgRNA activity by Cre recombination for sequential editing of two loci.
- Author
-
Chylinski K, Hubmann M, Hanna RE, Yanchus C, Michlits G, Uijttewaal ECH, Doench J, Schramek D, and Elling U
- Subjects
- Animals, CRISPR-Cas Systems, Clustered Regularly Interspaced Short Palindromic Repeats, Genetic Engineering, Homologous Recombination, Integrases, Mice, Mutagenesis, RNA Polymerase III, Gene Editing methods, Mouse Embryonic Stem Cells metabolism, RNA, Guide, CRISPR-Cas Systems genetics
- Abstract
CRISPR-Cas9 is an efficient and versatile tool for genome engineering in many species. However, inducible CRISPR-Cas9 editing systems that regulate Cas9 activity or sgRNA expression often suffer from significant limitations, including reduced editing capacity, off-target effects, or leaky expression. Here, we develop a precisely controlled sgRNA expression cassette that can be combined with widely-used Cre systems, termed CRISPR-Switch (SgRNA With Induction/Termination by Cre Homologous recombination). Switch-ON facilitates controlled, rapid induction of sgRNA activity. In turn, Switch-OFF-mediated termination of editing improves generation of heterozygous genotypes and can limit off-target effects. Furthermore, we design sequential CRISPR-Switch-based editing of two loci in a strictly programmable manner and determined the order of mutagenic events that leads to development of glioblastoma in mice. Thus, CRISPR-Switch substantially increases the versatility of gene editing through precise and rapid switching ON or OFF sgRNA activity, as well as switching OVER to secondary sgRNAs.
- Published
- 2019
- Full Text
- View/download PDF
47. Toronto Workshop on Late Recurrence in Estrogen Receptor-Positive Breast Cancer: Part 1: Late Recurrence: Current Understanding, Clinical Considerations.
- Author
-
Dowling RJO, Kalinsky K, Hayes DF, Bidard FC, Cescon DW, Chandarlapaty S, Deasy JO, Dowsett M, Gray RJ, Henry NL, Meric-Bernstam F, Perlmutter J, Sledge GW, Bratman SV, Carey LA, Chang MC, DeMichele A, Ennis M, Jerzak KJ, Korde LA, Lohmann AE, Mamounas EP, Parulekar WR, Regan MM, Schramek D, Stambolic V, Thorat MA, Whelan TJ, Wolff AC, Woodgett JR, Sparano JA, and Goodwin PJ
- Abstract
Disease recurrence (locoregional, distant) exerts a significant clinical impact on the survival of estrogen receptor-positive breast cancer patients. Many of these recurrences occur late, more than 5 years after original diagnosis, and represent a major obstacle to the effective treatment of this disease. Indeed, methods to identify patients at risk of late recurrence and therapeutic strategies designed to avert or treat these recurrences are lacking. Therefore, an international workshop was convened in Toronto, Canada, in February 2018 to review the current understanding of late recurrence and to identify critical issues that require future study. In this article, the major issues surrounding late recurrence are defined and current approaches that may be applicable to this challenge are discussed. Specifically, diagnostic tests with potential utility in late-recurrence prediction are described as well as a variety of patient-related factors that may influence recurrence risk. Clinical and therapeutic approaches are also reviewed, with a focus on patient surveillance and the implementation of extended endocrine therapy in the context of late-recurrence prevention. Understanding and treating late recurrence in estrogen receptor-positive breast cancer is a major unmet clinical need. A concerted effort of basic and clinical research is required to confront late recurrence and improve disease management and patient survival., (© The Author(s) 2019. Published by Oxford University Press.)
- Published
- 2019
- Full Text
- View/download PDF
48. Toronto Workshop on Late Recurrence in Estrogen Receptor-Positive Breast Cancer: Part 2: Approaches to Predict and Identify Late Recurrence, Research Directions.
- Author
-
Dowling RJO, Sparano JA, Goodwin PJ, Bidard FC, Cescon DW, Chandarlapaty S, Deasy JO, Dowsett M, Gray RJ, Henry NL, Meric-Bernstam F, Perlmutter J, Sledge GW, Thorat MA, Bratman SV, Carey LA, Chang MC, DeMichele A, Ennis M, Jerzak KJ, Korde LA, Lohmann AE, Mamounas EP, Parulekar WR, Regan MM, Schramek D, Stambolic V, Whelan TJ, Wolff AC, Woodgett JR, Kalinsky K, and Hayes DF
- Abstract
Late disease recurrence (more than 5 years after initial diagnosis) represents a clinical challenge in the treatment and management of estrogen receptor-positive breast cancer (BC). An international workshop was convened in Toronto, Canada, in February 2018 to review the current understanding of late recurrence and to identify critical issues that require future study. The underlying biological causes of late recurrence are complex, with the processes governing cancer cell dormancy, including immunosurveillance, cell proliferation, angiogenesis, and cellular stemness, being integral to disease progression. These critical processes are described herein as well as their role in influencing risk of recurrence. Moreover, observational and interventional clinical trials are proposed, with a focus on methods to identify patients at risk of recurrence and possible strategies to combat this in patients with estrogen receptor-positive BC. Because the problem of late BC recurrence of great importance, recent advances in disease detection and patient monitoring should be incorporated into novel clinical trials to evaluate approaches to enhance patient management. Indeed, future research on these issues is planned and will offer new options for effective late recurrence treatment and prevention strategies., (© The Author(s) 2019. Published by Oxford University Press.)
- Published
- 2019
- Full Text
- View/download PDF
49. Apelin inhibition prevents resistance and metastasis associated with anti-angiogenic therapy.
- Author
-
Uribesalgo I, Hoffmann D, Zhang Y, Kavirayani A, Lazovic J, Berta J, Novatchkova M, Pai TP, Wimmer RA, László V, Schramek D, Karim R, Tortola L, Deswal S, Haas L, Zuber J, Szűcs M, Kuba K, Dome B, Cao Y, Haubner BJ, and Penninger JM
- Subjects
- Animals, Apelin antagonists & inhibitors, Apelin deficiency, Apelin genetics, Apelin Receptors antagonists & inhibitors, Apelin Receptors deficiency, Apelin Receptors genetics, Cell Line, Tumor, Endothelial Cells drug effects, Endothelial Cells metabolism, Endothelial Cells pathology, Female, Gene Expression Regulation, Neoplastic, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mouse Embryonic Stem Cells drug effects, Mouse Embryonic Stem Cells metabolism, Mouse Embryonic Stem Cells pathology, Neoplasm Metastasis, Signal Transduction, Tumor Burden drug effects, Tumor Microenvironment, Angiogenesis Inhibitors pharmacology, Apelin metabolism, Apelin Receptors metabolism, Cell Movement drug effects, Drug Resistance, Neoplasm, Lung Neoplasms drug therapy, Mammary Neoplasms, Experimental drug therapy, Neovascularization, Pathologic, Protein Kinase Inhibitors pharmacology, Sunitinib pharmacology
- Abstract
Angiogenesis is a hallmark of cancer, promoting growth and metastasis. Anti-angiogenic treatment has limited efficacy due to therapy-induced blood vessel alterations, often followed by local hypoxia, tumor adaptation, progression, and metastasis. It is therefore paramount to overcome therapy-induced resistance. We show that Apelin inhibition potently remodels the tumor microenvironment, reducing angiogenesis, and effectively blunting tumor growth. Functionally, targeting Apelin improves vessel function and reduces polymorphonuclear myeloid-derived suppressor cell infiltration. Importantly, in mammary and lung cancer, Apelin prevents resistance to anti-angiogenic receptor tyrosine kinase (RTK) inhibitor therapy, reducing growth and angiogenesis in lung and breast cancer models without increased hypoxia in the tumor microenvironment. Apelin blockage also prevents RTK inhibitor-induced metastases, and high Apelin levels correlate with poor prognosis of anti-angiogenic therapy patients. These data identify a druggable anti-angiogenic drug target that reduces tumor blood vessel densities and normalizes the tumor vasculature to decrease metastases., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
50. AIF-regulated oxidative phosphorylation supports lung cancer development.
- Author
-
Rao S, Mondragón L, Pranjic B, Hanada T, Stoll G, Köcher T, Zhang P, Jais A, Lercher A, Bergthaler A, Schramek D, Haigh K, Sica V, Leduc M, Modjtahedi N, Pai TP, Onji M, Uribesalgo I, Hanada R, Kozieradzki I, Koglgruber R, Cronin SJ, She Z, Quehenberger F, Popper H, Kenner L, Haigh JJ, Kepp O, Rak M, Cai K, Kroemer G, and Penninger JM
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Datasets as Topic, Disease Progression, Glycolysis, Humans, Lung Neoplasms pathology, Mice, Mice, Inbred C57BL, Oxidative Phosphorylation, Apoptosis Inducing Factor physiology, Carcinogenesis metabolism, Carcinoma, Non-Small-Cell Lung metabolism, Lung Neoplasms metabolism
- Abstract
Cancer is a major and still increasing cause of death in humans. Most cancer cells have a fundamentally different metabolic profile from that of normal tissue. This shift away from mitochondrial ATP synthesis via oxidative phosphorylation towards a high rate of glycolysis, termed Warburg effect, has long been recognized as a paradigmatic hallmark of cancer, supporting the increased biosynthetic demands of tumor cells. Here we show that deletion of apoptosis-inducing factor (AIF) in a Kras
G12D -driven mouse lung cancer model resulted in a marked survival advantage, with delayed tumor onset and decreased malignant progression. Mechanistically, Aif deletion leads to oxidative phosphorylation (OXPHOS) deficiency and a switch in cellular metabolism towards glycolysis in non-transformed pneumocytes and at early stages of tumor development. Paradoxically, although Aif-deficient cells exhibited a metabolic Warburg profile, this bioenergetic change resulted in a growth disadvantage of KrasG12D -driven as well as Kras wild-type lung cancer cells. Cell-autonomous re-expression of both wild-type and mutant AIF (displaying an intact mitochondrial, but abrogated apoptotic function) in Aif-knockout KrasG12D mice restored OXPHOS and reduced animal survival to the same level as AIF wild-type mice. In patients with non-small cell lung cancer, high AIF expression was associated with poor prognosis. These data show that AIF-regulated mitochondrial respiration and OXPHOS drive the progression of lung cancer.- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.