ASCL1 is activated downstream of the ROR2/CREB signaling pathway to support lineage plasticity in prostate cancer.
暂无分享,去创建一个
A. Zoubeidi | Dwaipayan Ganguli | M. Kobelev | O. Sivak | Amina Talal | Nakisa Tabrizian | Takeshi Namekawa | Shaghayegh Nouruzi | Cassandra Jingjing Cui | Ishana Lodhia | C. Cui
[1] S. Balk,et al. WNT5a Signaling through ROR2 Activates the Hippo Pathway to Suppress YAP1 Activity and Tumor Growth. , 2023, Cancer research.
[2] P. Lopez-Bergami. ROR2, a driver of “phenotype switching” in melanoma? , 2022, Cancer Cell International.
[3] Tao Wang,et al. Ectopic JAK–STAT activation enables the transition to a stem-like and multilineage state conferring AR-targeted therapy resistance , 2022, Nature Cancer.
[4] L. Mazutis,et al. Lineage plasticity in prostate cancer depends on JAK/STAT inflammatory signaling , 2022, Science.
[5] A. Yu,et al. Bioengineered BERA-Wnt5a siRNA targeting Wnt5a/FZD2 signaling suppresses advanced prostate cancer tumor growth and enhances enzalutamide treatment. , 2022, Molecular cancer therapeutics.
[6] Wen-Hui Lien,et al. ROR2 regulates self-renewal and maintenance of hair follicle stem cells , 2022, Nature Communications.
[7] A. Zoubeidi,et al. ASCL1 activates neuronal stem cell-like lineage programming through remodeling of the chromatin landscape in prostate cancer , 2022, Nature Communications.
[8] P. Lopez-Bergami,et al. Cellular and Molecular Mechanisms Implicated in the Dual Role of ROR2 in Cancer. , 2022, Critical reviews in oncology/hematology.
[9] H. G. van der Poel,et al. Drug-induced epigenomic plasticity reprograms circadian rhythm regulation to drive prostate cancer towards androgen-independence , 2021, medRxiv.
[10] H. G. van der Poel,et al. An androgen receptor switch underlies lineage infidelity in treatment-resistant prostate cancer , 2021, Nature Cell Biology.
[11] Jie Wang,et al. Non-apoptotic function of caspase-8 confers prostate cancer enzalutamide resistance via NF-κB activation , 2021, Cell Death & Disease.
[12] V. Quaranta,et al. ASCL1 represses a SOX9+ neural crest stem-like state in small cell lung cancer , 2021, Genes & development.
[13] Liewei Wang,et al. A noncanonical AR addiction drives enzalutamide resistance in prostate cancer , 2021, Nature Communications.
[14] A. Bleckmann,et al. The WNT/ROR Pathway in Cancer: From Signaling to Therapeutic Intervention , 2021, Cells.
[15] A. Gao,et al. WLS-Wnt signaling promotes neuroendocrine prostate cancer , 2021, iScience.
[16] M. Rubin,et al. Impact of Lineage Plasticity to and from a Neuroendocrine Phenotype on Progression and Response in Prostate and Lung Cancers. , 2020, Molecular cell.
[17] Oh-Joon Kwon,et al. Sox2 is necessary for androgen ablation-induced neuroendocrine differentiation from Pten null Sca-1+ prostate luminal cells , 2020, Oncogene.
[18] Wen-en Zhao,et al. Current development of CBP/p300 inhibitors in the last decade. , 2020, European journal of medicinal chemistry.
[19] P. Lopez-Bergami,et al. The emerging role of Wnt5a in the promotion of a pro-inflammatory and immunosuppressive tumor microenvironment , 2020, Cancer and Metastasis Reviews.
[20] Haiyong Peng,et al. Affinity maturation, humanization, and co-crystallization of a rabbit anti-human ROR2 monoclonal antibody for therapeutic applications , 2020, The Journal of Biological Chemistry.
[21] M. Rubin,et al. Role of specialized composition of SWI/SNF complexes in prostate cancer lineage plasticity , 2020, Nature Communications.
[22] F. D. de Sauvage,et al. The great escape: tumour cell plasticity in resistance to targeted therapy , 2019, Nature Reviews Drug Discovery.
[23] P. Nelson,et al. Molecular profiling stratifies diverse phenotypes of treatment-refractory metastatic castration-resistant prostate cancer. , 2019, The Journal of clinical investigation.
[24] G. Patel,et al. Neuroendocrine Differentiation of Prostate Cancer—An Intriguing Example of Tumor Evolution at Play , 2019, Cancers.
[25] M. Loda,et al. The Role of Lineage Plasticity in Prostate Cancer Therapy Resistance , 2019, Clinical Cancer Research.
[26] A. Villers,et al. ARCHES: A Randomized, Phase III Study of Androgen Deprivation Therapy With Enzalutamide or Placebo in Men With Metastatic Hormone-Sensitive Prostate Cancer , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[27] B. Biswal,et al. Cell signaling and cancer: a mechanistic insight into drug resistance , 2019, Molecular Biology Reports.
[28] L. Mei,et al. Agrin-Lrp4-Ror2 signaling regulates adult hippocampal neurogenesis in mice , 2019, eLife.
[29] M. Stockler,et al. Enzalutamide with Standard First-Line Therapy in Metastatic Prostate Cancer. , 2019, The New England journal of medicine.
[30] N. Agarwal,et al. Apalutamide for Metastatic, Castration-Sensitive Prostate Cancer. , 2019, The New England journal of medicine.
[31] Yi Mi Wu,et al. Genomic correlates of clinical outcome in advanced prostate cancer , 2019, Proceedings of the National Academy of Sciences.
[32] R. Kageyama,et al. High Hes1 expression and resultant Ascl1 suppression regulate quiescent vs. active neural stem cells in the adult mouse brain , 2019, Genes & development.
[33] Steven L. Chang,et al. Evaluation of Intense Androgen Deprivation Before Prostatectomy: A Randomized Phase II Trial of Enzalutamide and Leuprolide With or Without Abiraterone. , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[34] Libing Song,et al. Wnt5a induces and maintains prostate cancer cells dormancy in bone , 2018, The Journal of experimental medicine.
[35] A. Falini,et al. The proneural gene ASCL1 governs the transcriptional subgroup affiliation in glioblastoma stem cells by directly repressing the mesenchymal gene NDRG1 , 2018, Cell Death & Differentiation.
[36] Xiaoyan Zhang,et al. Cistrome Data Browser: expanded datasets and new tools for gene regulatory analysis , 2018, Nucleic Acids Res..
[37] R. Beerli,et al. Novel Antibody Drug Conjugates Targeting Tumor-Associated Receptor Tyrosine Kinase ROR2 by Functional Screening of Fully Human Antibody Libraries Using Transpo-mAb Display on Progenitor B Cells , 2018, Front. Immunol..
[38] M. Ohba,et al. Receptor Tyrosine Kinase-Targeted Cancer Therapy , 2018, International journal of molecular sciences.
[39] Zheng Wang,et al. Androgen deprivation promotes neuroendocrine differentiation and angiogenesis through CREB-EZH2-TSP1 pathway in prostate cancers , 2018, Nature Communications.
[40] Joshua M. Stuart,et al. Clinical and Genomic Characterization of Treatment-Emergent Small-Cell Neuroendocrine Prostate Cancer: A Multi-institutional Prospective Study. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[41] F. Saad,et al. Enzalutamide in Men with Nonmetastatic, Castration‐Resistant Prostate Cancer , 2018, The New England journal of medicine.
[42] M. Rubin,et al. Patient derived organoids to model rare prostate cancer phenotypes , 2018, Nature Communications.
[43] G. Hampton,et al. A transcriptional MAPK Pathway Activity Score (MPAS) is a clinically relevant biomarker in multiple cancer types , 2018, npj Precision Oncology.
[44] A. Zoubeidi,et al. Cellular plasticity and the neuroendocrine phenotype in prostate cancer , 2018, Nature Reviews Urology.
[45] Y. Minami,et al. Diverse roles for the ror‐family receptor tyrosine kinases in neurons and glial cells during development and repair of the nervous system , 2018, Developmental dynamics : an official publication of the American Association of Anatomists.
[46] Michael D. Nyquist,et al. Androgen Receptor Pathway-Independent Prostate Cancer Is Sustained through FGF Signaling. , 2017, Cancer cell.
[47] Gary D Bader,et al. ASCL1 Reorganizes Chromatin to Direct Neuronal Fate and Suppress Tumorigenicity of Glioblastoma Stem Cells. , 2017, Cell stem cell.
[48] M. Rubin,et al. Transdifferentiation as a Mechanism of Treatment Resistance in a Mouse Model of Castration-Resistant Prostate Cancer. , 2017, Cancer discovery.
[49] V. Njar,et al. Dissecting major signaling pathways in prostate cancer development and progression: Mechanisms and novel therapeutic targets , 2017, The Journal of Steroid Biochemistry and Molecular Biology.
[50] Ximing J. Yang,et al. FOXA1 inhibits prostate cancer neuroendocrine differentiation , 2017, Oncogene.
[51] Henry W. Long,et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance , 2017, Science.
[52] M. Rubin,et al. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer , 2017, Science.
[53] A. Aiba,et al. Critical role of Ror2 receptor tyrosine kinase in regulating cell cycle progression of reactive astrocytes following brain injury , 2017, Glia.
[54] Tao Liu,et al. Cistrome Data Browser: a data portal for ChIP-Seq and chromatin accessibility data in human and mouse , 2016, Nucleic Acids Res..
[55] Hedi Peterson,et al. g:Profiler—a web server for functional interpretation of gene lists (2016 update) , 2016, Nucleic Acids Res..
[56] Fidel Ramírez,et al. deepTools2: a next generation web server for deep-sequencing data analysis , 2016, Nucleic Acids Res..
[57] Matteo Benelli,et al. Divergent clonal evolution of castration resistant neuroendocrine prostate cancer , 2016, Nature Medicine.
[58] Sridhar Ramaswamy,et al. RNA-Seq of single prostate CTCs implicates noncanonical Wnt signaling in antiandrogen resistance , 2015, Science.
[59] Y. Ohe,et al. Expression profiling of receptor tyrosine kinases in high-grade neuroendocrine carcinoma of the lung: a comparative analysis with adenocarcinoma and squamous cell carcinoma , 2015, Journal of Cancer Research and Clinical Oncology.
[60] Yezi Zhu,et al. Intracrine Androgens and AKR1C3 Activation Confer Resistance to Enzalutamide in Prostate Cancer. , 2015, Cancer research.
[61] Wei Li,et al. Agonist and antagonist switch DNA motifs recognized by human androgen receptor in prostate cancer , 2015, The EMBO journal.
[62] Bin Zhang,et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations , 2014, Nucleic Acids Res..
[63] Zhaohui S. Qin,et al. Therapeutic Targeting of BET Bromodomain Proteins in Castration-Resistant Prostate Cancer , 2014, Nature.
[64] Susan S. Taylor,et al. Kinases and Pseudokinases: Lessons from RAF , 2014, Molecular and Cellular Biology.
[65] M. Rubin,et al. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. , 2014, Cancer research.
[66] Irena Nowak,et al. Erk/MAP Kinase Signaling Pathway and Neuroendocrine Differentiation of Non–Small-Cell Lung Cancer , 2014, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.
[67] M. Lemmon,et al. Receptor tyrosine kinases with intracellular pseudokinase domains. , 2013, Biochemical Society transactions.
[68] M. Gleave,et al. A Novel Antiandrogen, Compound 30, Suppresses Castration-Resistant and MDV3100-Resistant Prostate Cancer Growth In Vitro and In Vivo , 2013, Molecular Cancer Therapeutics.
[69] Kurt Miller,et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. , 2012, The New England journal of medicine.
[70] Benjamin E. Gross,et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. , 2012, Cancer discovery.
[71] Y. Minami,et al. Ror family receptor tyrosine kinases regulate the maintenance of neural progenitor cells in the developing neocortex , 2012, Journal of Cell Science.
[72] David R. Kelley,et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks , 2012, Nature Protocols.
[73] M. Greenberg,et al. Wnt5a–Ror–Dishevelled signaling constitutes a core developmental pathway that controls tissue morphogenesis , 2012, Proceedings of the National Academy of Sciences.
[74] Arturo Molina,et al. Abiraterone and increased survival in metastatic prostate cancer. , 2011, The New England journal of medicine.
[75] P. Pujol,et al. ROR2 Promoter Methylation Change in Osteoblastic Differentiation of Mesenchymal Stem Cells , 2011, Cell journal.
[76] D. V. van Aalten,et al. Pseudokinases-remnants of evolution or key allosteric regulators? , 2010, Current opinion in structural biology.
[77] Y. Minami,et al. Ror2/Frizzled Complex Mediates Wnt5a-Induced AP-1 Activation by Regulating Dishevelled Polymerization , 2010, Molecular and Cellular Biology.
[78] J. Humm,et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1–2 study , 2010, The Lancet.
[79] N. Oue,et al. Wnt5a signaling is involved in the aggressiveness of prostate cancer and expression of metalloproteinase , 2010, Oncogene.
[80] Gonçalo R. Abecasis,et al. The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..
[81] Mitch Dowsett,et al. Phase I clinical trial of a selective inhibitor of CYP17, abiraterone acetate, confirms that castration-resistant prostate cancer commonly remains hormone driven. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[82] Clifford A. Meyer,et al. Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.
[83] D. Gold,et al. Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation , 2008, Nature Genetics.
[84] H. Frierson,et al. Androgen-independent growth and tumorigenesis of prostate cancer cells are enhanced by the presence of PKA-differentiated neuroendocrine cells. , 2007, Cancer research.
[85] G. Barton,et al. Emerging roles of pseudokinases. , 2006, Trends in cell biology.
[86] T. Golub,et al. Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. , 2006, Cancer research.
[87] Pablo Tamayo,et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.
[88] T. Hunter,et al. The mouse kinome: discovery and comparative genomics of all mouse protein kinases. , 2004, Proceedings of the National Academy of Sciences of the United States of America.
[89] M. Daly,et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes , 2003, Nature Genetics.
[90] T. Hunter,et al. The Protein Kinase Complement of the Human Genome , 2002, Science.
[91] Jayoung Kim,et al. Activation of the Erk mitogen-activated protein kinase pathway stimulates neuroendocrine differentiation in LNCaP cells independently of cell cycle withdrawal and STAT3 phosphorylation. , 2002, Cancer research.
[92] D. Robinson,et al. The protein tyrosine kinase family of the human genome , 2000, Oncogene.
[93] M. Dragunow,et al. Is CREB a key to neuronal survival? , 2000, Trends in Neurosciences.
[94] E. Perens,et al. A C. elegans Ror receptor tyrosine kinase regulates cell motility and asymmetric cell division , 1999, Nature.
[95] D. Alessi,et al. Mitogen‐ and stress‐activated protein kinase‐1 (MSK1) is directly activated by MAPK and SAPK2/p38, and may mediate activation of CREB , 1998, The EMBO journal.
[96] P. Masiakowski,et al. A novel family of cell surface receptors with tyrosine kinase-like domain. , 1992, The Journal of biological chemistry.
[97] Joseph Schlessinger,et al. Signal transduction by receptors with tyrosine kinase activity , 1990, Cell.
[98] M. Montminy,et al. Cyclic AMP stimulates somatostatin gene transcription by phosphorylation of CREB at serine 133 , 1989, Cell.
[99] M. Rubin,et al. The Master Neural Transcription Factor BRN2 Is an Androgen Receptor-Suppressed Driver of Neuroendocrine Differentiation in Prostate Cancer. , 2017, Cancer discovery.
[100] Thomas R. Gingeras,et al. STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..
[101] Claude-Alain H. Roten,et al. Fast and accurate short read alignment with Burrows–Wheeler transform , 2009, Bioinform..