Mutational Inactivation of Mtorc1 Repressor Gene DEPDC5 in Human Gastrointestinal Stromal Tumors

Total Page:16

File Type:pdf, Size:1020Kb

Mutational Inactivation of Mtorc1 Repressor Gene DEPDC5 in Human Gastrointestinal Stromal Tumors Mutational inactivation of mTORC1 repressor gene DEPDC5 in human gastrointestinal stromal tumors Yuzhi Panga,1, Feifei Xiea,1, Hui Caob,1, Chunmeng Wangc,d,1, Meijun Zhue, Xiaoxiao Liua, Xiaojing Lua, Tao Huangf, Yanying Sheng,KeLia, Xiaona Jiaa, Zhang Lia, Xufen Zhenga, Simin Wanga,YiHeh, Linhui Wangi, Jonathan A. Fletchere,2, and Yuexiang Wanga,2 aKey Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences–Changzheng Hospital Joint Center for Translational Medicine, Changzheng Hospital, Institutes for Translational Medicine, Chinese Academy of Sciences–Second Military Medical University, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031 Shanghai, China; bDepartment of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127 Shanghai, China; cDepartment of Bone and Soft Tissue Sarcomas, Fudan University Shanghai Cancer Center, 200032 Shanghai, China; dDepartment of Oncology, Shanghai Medical College, Fudan University, 200032 Shanghai, China; eDepartment of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115; fShanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China; gDepartment of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127 Shanghai, China; hDepartment of Urology, No.1 Hospital of Jiaxing, 314000 Jiaxing, China; and iDepartment of Urology, Changzheng Hospital, Second Military Medical University, 200003 Shanghai, China Edited by Kenneth W. Kinzler, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, and approved October 1, 2019 (received for review August 26, 2019) Gastrointestinal stromal tumors (GISTs) are the most common demonstrate that chromosome 22q-targeting DEPDC5, silenced human sarcoma and are initiated by activating mutations in the by somatic mutations, is a GIST specific tumor suppressor and a KIT or PDGFRA receptor tyrosine kinases. Chromosome 22q dele- TKI treatment response modifier. tions are well-recognized frequent abnormalities in GISTs, occur- ring in ∼50% of GISTs. These deletions are thought to contribute Results and Discussion to the pathogenesis of this disease via currently unidentified tu- Whole Exome Sequencing Identifies Recurrent Inactivating DEPDC5 mor suppressor mechanisms. Using whole exome sequencing, we Aberrations in GISTs. To identify the causative tumor suppressor DEPDC5 report recurrent genomic inactivated gene mutations in genes at chromosome 22q in GISTs, we performed whole exome GISTs (16.4%, 9 of 55 patients). The demonstration of clonal sequencing in 40 GIST patients (Dataset S1). These studies con- DEPDC5 inactivation mutations in longitudinal specimens and in firmed reported GIST genes, such as KIT (3), PDGFRA (4), RB1 multiple metastases from individual patients suggests that these CDKN2A DMD MAX SETD2 mutations have tumorigenic roles in GIST progression. DEPDC5 in- (15), (15), (16), (17), and (18) activation promotes GIST tumor growth in vitro and in nude mice. (Datasets S2 and S3). Notably, these studies revealed somatic ho- DEPDC5 reduces cell proliferation through the mTORC1-signaling mozygous inactivating genomic DEPDC5 (encoding Dishevelled, Egl- pathway and subsequently induces cell-cycle arrest. Furthermore, 10 and Pleckstrin [DEP] domain-containing protein 5) aberrations, DEPDC5 modulates the sensitivity of GIST to KIT inhibitors, and the combination therapy with mTOR inhibitor and KIT inhibitor may Significance work better in GIST patients with DEPDC5 inactivation. These find- ings of recurrent genomic alterations, together with functional Activating mutations of KIT or PDGFRA are initiating events in data, validate the DEPDC5 as a bona fide tumor suppressor con- most gastrointestinal stromal tumors (GISTs) and indeed are tributing to GIST progression and a biologically relevant target of present in micro-GISTs, which are asymptomatic subcentimeter the frequent chromosome 22q deletions. lesions found in one-third of the general population. The bio- logical underpinnings of GIST progression are poorly un- sarcoma | GIST | KIT tyrosine kinase inhibitors | DEPDC5 derstood. Chromosome 22q deletions are well-recognized abnormalities in GISTs. However, the crucial gene has been arcomas are diverse mesenchymal malignancies that account unknown. We report recurrent genomic inactivated DEPDC5 Sfor ∼20% of pediatric and 1% of adult cancers (1). Gastro- mutations in GISTs. The DEPDC5 inactivated mutations are intestinal stromal tumors (GISTs) are the most common human prognostic in that they are associated with aggressive GISTs in sarcoma (2), which are mostly initiated by activating mutations of which they promote GIST progression and reduce sensitivity to the receptor tyrosine kinase KIT (75–80%) or PDGFRA (5–10%) KIT inhibitors. (3, 4). Although sharing the same KIT/PDGFRA mutations, micro- GISTs have a limited growth potential and hence are restrained at Author contributions: J.A.F. and Y.W. designed research; Y.P., F.X., H.C., C.W., M.Z., X. Liu, the subcentimeter level. The fact that micro-GISTs are common in X. Lu, Y.S., K.L., X.J., Z.L., X.Z., S.W., and Y.W. performed research; Y.P., F.X., H.C., C.W., and J.A.F. provided samples and clinical data; Y.P., F.X., C.W., M.Z., X. Liu, X. Lu, T.H., Y.S., general individuals (found in one-third of the general population) K.L., X.J., Z.L., X.Z., S.W., Y.H., L.W., J.A.F., and Y.W. analyzed data; and Y.P. and Y.W. without clinical symptoms (5–7) indicates that additional genetic wrote the paper. alterations contribute to the progression of clinical GISTs. Chro- The authors declare no competing interest. mosome 22q deletions are frequent chromosomal abnormalities in This article is a PNAS Direct Submission. ∼ – human GISTs, occurring in 50% of GISTs (2, 8 11), and are Published under the PNAS license. thought to contribute to the pathogenesis of this disease by yet- – Data deposition: Whole exome sequencing and RNA sequencing data reported in this unidentified tumor suppressor mechanisms (2, 8 11). study have been deposited in the National Omics Data Encyclopedia (accession no. Most GISTs with activating mutations in KIT often respond to OEP000478). treatment with KIT tyrosine kinase inhibitors (TKIs), such as first- 1Y.P., F.X., H.C., and C.W. contributed equally to this work. line imatinib, second-line sunitinib and third-line regorafenib, but 2To whom correspondence may be addressed. Email: [email protected] or the magnitude of tumor regression is variable (12–14). This het- [email protected]. erogeneity in TKI response could result from genetic modifiers This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. that regulate the degree to which tumor cells are dependent upon 1073/pnas.1914542116/-/DCSupplemental. the driver kinase and the response to TKI treatment. Here we First published October 21, 2019. 22746–22753 | PNAS | November 5, 2019 | vol. 116 | no. 45 www.pnas.org/cgi/doi/10.1073/pnas.1914542116 Downloaded by guest on September 30, 2021 including nonsense mutation, frameshift mutation, and dele- have somatic homozygous DEPDC5-inactivating aberrations tions in 7 of 40 (17.5%) GIST patients (Figs. 1 A and B and Dataset in 9 GIST patients (16.4%). Of 55 patients, 31 (∼56%) har- S1). Homozygous DEPDC5 mutations were confirmed by Sanger bored chromosome 22 loss (Dataset S4). All of the 9 patients sequencing (SI Appendix,Fig.S1A), single-nucleotide polymorphism harboring genomic DEPDC5 aberrations contain chromosome 22 (SNP) arrays (SI Appendix,Fig.S1B), quantitative PCR (SI Appendix, loss (Dataset S4). Therefore, DEPDC5 aberrations are significantly Fig. S1C), and fluorescence in situ hybridization (SI Appendix,Fig. more frequent in GISTs with chromosome 22 loss compared to S1D). Somatic inactivating genomic DEPDC5 aberrations were vali- chromosome 22 normal copy number (29 vs. 0%, P = 0.01177, 2- dated in 2 of 15 (13.3%) additional GIST patients (cases 41 to 55, tailed Fisher’stest)(Dataset S4). All of the 9 patients with genomic Dataset S4). This total set of 55 GIST patients was shown to DEPDC5 aberrations have both copies of DEPCD5 inactivated MEDICAL SCIENCES Fig. 1. Genomic DEPDC5 aberrations in 40 GIST patients. (A) Whole exome sequencing identifies genomic DEPDC5 aberrations in 7 of 40 (17.5%) GIST patients. Inactivating DEPDC5 mutations were intragenic homozygous deletions (blue lines indicate deleted exons) and hemizygous nucleotide alterations. Mutations are described according to international guidelines for sequence variant nomenclature provided by the Human Genome Variation Society (http:// varnomen.hgvs.org). Annotations in blue represent the nucleotide coding sequence mutations (indicated by “c.”) whereas annotations in green represent the resultant protein sequence mutations (indicated by “p.”). (B) Integrative genome viewer images of part of chromosome 22q from matched tumor and nonneoplastic cell DNAs from the same patients, demonstrating the tumor-restricted nature of DEPDC5 mutations. Pang et al. PNAS | November 5, 2019 | vol. 116 | no. 45 | 22747 Downloaded by guest on September 30, 2021 Fig. 2. Genomic DEPDC5 aberrations are a clonal genetic event in GIST progression. (A) Multiple tumors from the same patients share the
Recommended publications
  • Discovery of Novel Putative Tumor Suppressors from CRISPR Screens Reveals Rewired 2 Lipid Metabolism in AML Cells 3 4 W
    bioRxiv preprint doi: https://doi.org/10.1101/2020.10.08.332023; this version posted August 20, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 Discovery of novel putative tumor suppressors from CRISPR screens reveals rewired 2 lipid metabolism in AML cells 3 4 W. Frank Lenoir1,2, Micaela Morgado2, Peter C DeWeirdt3, Megan McLaughlin1,2, Audrey L 5 Griffith3, Annabel K Sangree3, Marissa N Feeley3, Nazanin Esmaeili Anvar1,2, Eiru Kim2, Lori L 6 Bertolet2, Medina Colic1,2, Merve Dede1,2, John G Doench3, Traver Hart2,4,* 7 8 9 1 - The University of Texas MD Anderson Cancer Center UTHealth Graduate School of 10 Biomedical Sciences; The University of Texas MD Anderson Cancer Center, Houston, TX 11 12 2 - Department of Bioinformatics and Computational Biology, The University of Texas MD 13 Anderson Cancer Center, Houston, TX, USA 14 15 3 - Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA 16 17 4 - Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, 18 Houston, TX, USA 19 20 21 22 23 * - Corresponding author: [email protected] 24 25 bioRxiv preprint doi: https://doi.org/10.1101/2020.10.08.332023; this version posted August 20, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Biology of Human Tumors Research Mir-409-3P/-5P Promotes Tumorigenesis, Epithelial-To-Mesenchymal Transition, and Bone Metastasis of Human Prostate Cancer
    Published OnlineFirst June 24, 2014; DOI: 10.1158/1078-0432.CCR-14-0305 Clinical Cancer Biology of Human Tumors Research miR-409-3p/-5p Promotes Tumorigenesis, Epithelial-to-Mesenchymal Transition, and Bone Metastasis of Human Prostate Cancer Sajni Josson1, Murali Gururajan1, Peizhen Hu1, Chen Shao1, Gina Chia-Yi Chu1, Haiyen E. Zhau1, Chunyan Liu1, Kaiqin Lao2, Chia-Lun Lu1, Yi-Tsung Lu1, Jake Lichterman1, Srinivas Nandana1, Quanlin Li3, Andre Rogatko3, Dror Berel3, Edwin M. Posadas1, Ladan Fazli4, Dhruv Sareen5, and Leland W.K. Chung1 Abstract Purpose: miR-409-3p/-5p is a miRNA expressed by embryonic stem cells, and its role in cancer biology and metastasis is unknown. Our pilot studies demonstrated elevated miR-409-3p/-5p expression in humanprostatecancerbonemetastaticcelllines; therefore, we defined the biologic impact of manipulation of miR-409-3p/-5p on prostate cancer progression and correlated the levels of its expression with clinical human prostate cancer bone metastatic specimens. Experimental Design: miRNA profiling of a prostate cancer bone metastatic epithelial-to-mesenchymal transition (EMT) cell line model was performed. A Gleason score human tissue array was probed for validation of specific miRNAs. In addition, genetic manipulation of miR-409-3p/-5p was performed to determine its role in tumor growth, EMT, and bone metastasis in mouse models. Results: Elevated expression of miR-409-3p/-5p was observed in bone metastatic prostate cancer cell lines and human prostate cancer tissues with higher Gleason scores. Elevated miR-409-3p expression levels correlated with progression-free survival of patients with prostate cancer. Orthotopic delivery of miR-409- 3p/-5p in the murine prostate gland induced tumors where the tumors expressed EMT and stemness markers.
    [Show full text]
  • Gene Ontology Functional Annotations and Pleiotropy
    Network based analysis of genetic disease associations Sarah Gilman Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy under the Executive Committee of the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2013 Sarah Gilman All Rights Reserved ABSTRACT Network based analysis of genetic disease associations Sarah Gilman Despite extensive efforts and many promising early findings, genome-wide association studies have explained only a small fraction of the genetic factors contributing to common human diseases. There are many theories about where this “missing heritability” might lie, but increasingly the prevailing view is that common variants, the target of GWAS, are not solely responsible for susceptibility to common diseases and a substantial portion of human disease risk will be found among rare variants. Relatively new, such variants have not been subject to purifying selection, and therefore may be particularly pertinent for neuropsychiatric disorders and other diseases with greatly reduced fecundity. Recently, several researchers have made great progress towards uncovering the genetics behind autism and schizophrenia. By sequencing families, they have found hundreds of de novo variants occurring only in affected individuals, both large structural copy number variants and single nucleotide variants. Despite studying large cohorts there has been little recurrence among the genes implicated suggesting that many hundreds of genes may underlie these complex phenotypes. The question
    [Show full text]
  • Functional Characterization of the Candidate Tumor Suppressor Gene NPRL2/G21 Located in 3P21.3C
    [CANCER RESEARCH 64, 6438–6443, September 15, 2004] Functional Characterization of the Candidate Tumor Suppressor Gene NPRL2/G21 Located in 3p21.3C Jingfeng Li,1 Fuli Wang,1 Klas Haraldson,1 Alexey Protopopov,1 Fuh-Mei Duh,2,3 Laura Geil,2,3 Igor Kuzmin,2,3 John D. Minna,4 Eric Stanbridge,5 Eleonora Braga,6 Vladimir I. Kashuba,1,7 George Klein,1 Michael I. Lerman,2 and Eugene R. Zabarovsky1,8 1Microbiology and Tumor Biology Center, Center for Genomics and Bioinformatics, Karolinska Institute, Stockholm, Sweden; 2Cancer-Causing Genes Section, Laboratory of Immunobiology, Center for Cancer Research, National Cancer Institute, Frederick, Maryland; 3Basic Research Program, SAIC-Frederick, Inc., Frederick, Maryland; 4Hamon Center for Therapeutic Oncology, Research, University of Texas Southwestern Medical Center, Dallas, Texas; 5Department of Microbiology and Molecular Genetics, College of Medicine, University of California at Irvine, Irvine, California; 6Russian State Genetics Center, Moscow, Russia; 7Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kiev, Ukraine; and 8Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia ABSTRACT accompanied by chromosome 3p homozygous deletions, is a charac- teristic feature of most major epithelial carcinomas, such as lung, Initial analysis identified the NPRL2/G21 gene located in 3p21.3C, the breast, cervical, oral cavity, ovary, and kidney (2, 3). These changes lung cancer region, as a strong candidate tumor suppressor gene. Here we indicate the involvement of multiple tumor suppressor genes. provide additional evidence of the tumor suppressor function of NPRL2/ G21. The gene has highly conserved homologs/orthologs ranging from We have performed a comprehensive deletion survey of 3p on more yeast to humans.
    [Show full text]
  • Concordance of Copy Number Loss and Down-Regulation of Tumor Suppressor Genes: a Pan-Cancer Study Min Zhao1 and Zhongming Zhao2,3,4,5*
    The Author(s) BMC Genomics 2016, 17(Suppl 7):532 DOI 10.1186/s12864-016-2904-y RESEARCH Open Access Concordance of copy number loss and down-regulation of tumor suppressor genes: a pan-cancer study Min Zhao1 and Zhongming Zhao2,3,4,5* From The International Conference on Intelligent Biology and Medicine (ICIBM) 2015 Indianapolis, IN, USA. 13-15 November 2015 Abstract Background: Tumor suppressor genes (TSGs) encode the guardian molecules to control cell growth. The genomic alteration of TSGs may cause tumorigenesis and promote cancer progression. So far, investigators have mainly studied the functional effects of somatic single nucleotide variants in TSGs. Copy number variation (CNV) is another important form of genetic variation, and is often involved in cancer biology and drug treatment, but studies of CNV in TSGs are less represented in literature. In addition, there is a lack of a combinatory analysis of gene expression and CNV in this important gene set. Such a study may provide more insights into the relationship between gene dosage and tumorigenesis. To meet this demand, we performed a systematic analysis of CNVs and gene expression in TSGs to provide a systematic view of CNV and gene expression change in TSGs in pan-cancer. Results: We identified 1170 TSGs with copy number gain or loss in 5846 tumor samples. Among them, 207 TSGs tended to have copy number loss (CNL), from which fifteen CNL hotspot regions were identified. The functional enrichment analysis revealed that the 207 TSGs were enriched in cancer-related pathways such as P53 signaling pathway and the P53 interactome.
    [Show full text]
  • Functional Genetic Analysis Request Form
    Functional Genetic Analysis Request Form Please complete the form as far as possible. Completed forms can be faxed to 31-10-7043200, or sent as an encrypted PDF to the following email address: [email protected] For questions or further information, please contact [email protected]; [email protected]; [email protected] Patient information: Surname: First (given) name / initials: Date of birth (dd-mm-yyyy): Address: Medical insurance policy number: Material (where applicable); Blood: Fibroblasts: Other (please specify): please mark as appropriate Date sample was taken (if applicable): Gene variant information (HGVS nomenclature): Gene Name Reference transcript accession Nucleotide change (predicted protein variant) number Sender information Name: Email: Department (if applicable): Institute: Address: City: Country: version: 3 22-10-2018 Pag. 1/2 Functional Genetic Analysis Request Form Type of analysis requested: Diagnostic testing of a variant of uncertain clinical significance (VUS) Direct functional assay in patient material (eg. blood, fibroblasts, other) Functional test requested (please specify): Ciliopathies (fibroblasts required) Structural test, tubulin staining for cilia size and number Hedgehog signaling test DNA-repair defects (fibroblasts required) Xeroderma pigmentosum (XP) Ataxia telangiectasia (AT) Cockayne syndrome(CS) Nijmegen Breakage syndrome (NBS) Trichothiodystrophy (TTD) Mechanistic target of rapamycin (mTOR) signaling pathway (TSC1, TSC2, DEPDC5, NPRL2, NPRL3, AKT1, AKT3, TBC1D7)
    [Show full text]
  • SEA You Later Alli-GATOR – a Dynamic Regulator of the TORC1 Stress Response Pathway Svetlana Dokudovskaya1,* and Michael P
    © 2015. Published by The Company of Biologists Ltd | Journal of Cell Science (2015) 128, 2219-2228 doi:10.1242/jcs.168922 COMMENTARY SEA you later alli-GATOR – a dynamic regulator of the TORC1 stress response pathway Svetlana Dokudovskaya1,* and Michael P. Rout2 ABSTRACT equivalent of the vacuole) (Betz and Hall, 2013). Vacuoles and Cells constantly adapt to various environmental changes and lysosomes are the storage and recycling depots of the cell. Among their stresses. The way in which nutrient and stress levels in a cell feed many roles, they mediate protein degradation, store amino acids, and back to control metabolism and growth are, unsurprisingly, extremely sequester small ions and polyphosphates (Li and Kane, 2009; Luzio – complex, as responding with great sensitivity and speed to the ‘feast et al., 2007). These organelles are also key players in autophagy a or famine, slack or stress’ status of its environment is a central goal for process in which cytosol and organelles are sequestered within double- any organism. The highly conserved target of rapamycin complex 1 membraned vesicles, which deliver their contents to the vacuole or (TORC1) controls eukaryotic cell growth and response to a variety of lysosomefordegradationorrecycling (Yang and Klionsky, 2009). The signals, including nutrients, hormones and stresses, and plays the lysosome also plays an active role in amino acid sensing through its + key role in the regulation of autophagy. A lot of attention has been paid proton pump, the vacuolar type H -ATPase (v-ATPase) (Zoncu et al., recently to the factors in this pathway functioning upstream of TORC1. 2011), and it is likely that the vacuole plays a similar role.
    [Show full text]
  • Nprl2 (NM 018879) Mouse Tagged ORF Clone Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for MR205929 Nprl2 (NM_018879) Mouse Tagged ORF Clone Product data: Product Type: Expression Plasmids Product Name: Nprl2 (NM_018879) Mouse Tagged ORF Clone Tag: Myc-DDK Symbol: Nprl2 Synonyms: 2810446G01Rik; G21; NPR2L; Tusc4 Vector: pCMV6-Entry (PS100001) E. coli Selection: Kanamycin (25 ug/mL) Cell Selection: Neomycin ORF Nucleotide >MR205929 ORF sequence Sequence: Red=Cloning site Blue=ORF Green=Tags(s) TTTTGTAATACGACTCACTATAGGGCGGCCGGGAATTCGTCGACTGGATCCGGTACCGAGGAGATCTGCC GCCGCGATCGCC ATGGGCAGCAGCTGCCGCATCGAATGTATATTCTTCAGCGAGTTCCACCCAACGCTGGGACCCAAAATTA CCTATCAGGTCCCTGAGGATTTCATCTCGCGAGAACTGTTTGACACGGTCCAGGTGTACATCATCACCAA GCCCGAGTTACAGAACAAGCTCATCACTGTCACAGCCATGGAGAAGAAGCTGATTGGCTGCCCCGTGTGC ATTGAACACAAGAAGTACAGCCGCAATGCTCTCCTCTTCAACCTGGGCTTTGTGTGTGACGCTCAGGCTA AGACTTGTGCCCTGGAACCCATCGTTAAAAAGCTGGCCGGCTACTTGACCACGCTGGAGCTTGAGAGCAG CTTTGTATCCAACGAGGAGAGCAAGCAGAAGCTGGTGCCCATCATGACCATCCTGCTGGAAGAGCTGAAT GCCTCCGGCCGCTGCACTCTGCCCATTGACGAATCCAACACCATCCACTTGAAGGTGATCGAGCAGCGGC CAGACCCTCCTGTTGCGCAGGAGTATGATGTGCCCGTCTTTACAAAGGACAAGGAGGATTTCTTCAGCTC GCAGTGGGACCTTACCACACAGCAGATCCTGCCCTACATTGACGGGTTTCGCCATGTCCAGAAGATCTCC GCTGAGGCAGACGTGGAACTCAACCTGGTGCGCATCGCCATTCAGAACCTGCTGTACTATGGTGTTGTGA CACTGGTATCCATCCTCCAGTACTCCAATGTGTATTGCCCAACACCAAAAGTCCAGGATCTGGTGGATGA CAAGTCCCTGCAGGAGGCATGTCTGTCCTACGTCACCAAGCAAGGACACAAAAGGGCCAGTCTCCGAGAT GTGTTCCAGCTATACTGCAGCCTGAGCCCTGGTACCACTGTGCGAGACCTCATCGGCCGCCACCCCCAGC
    [Show full text]
  • Hdl 124036.Pdf
    Neurobiology of Disease 134 (2020) 104640 Contents lists available at ScienceDirect Neurobiology of Disease journal homepage: www.elsevier.com/locate/ynbdi Functional screening of GATOR1 complex variants reveals a role for mTORC1 deregulation in FCD and focal epilepsy T Ruby E. Dawsona,c, Alvaro F. Nieto Guilb,c, Louise J. Robertsonb,c, Sandra G. Piltzb,c, ⁎ James N. Hughesa,c, Paul Q. Thomasb,c,d, a School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia b School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia c Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia d Precision Medicine Theme, South Australia Health and Medical Research Institute, Adelaide, SA 5000, Australia ARTICLE INFO ABSTRACT Keywords: Mutations in the GAP activity toward RAGs 1 (GATOR1) complex genes (DEPDC5, NPRL2 and NPRL3) have been Neurodevelopment associated with focal epilepsy and focal cortical dysplasia (FCD). GATOR1 functions as an inhibitor of the Epilepsy mTORC1 signalling pathway, indicating that the downstream effects of mTORC1 deregulation underpin the Focal cortical dysplasia disease. However, the vast majority of putative disease-causing variants have not been functionally assessed for Molecular genetics mTORC1 repression activity. Here, we develop a novel in vitro functional assay that enables rapid assessment of Functional testing GATOR1-gene variants. Surprisingly, of the 17 variants tested, we show that only six showed significantly im- Developmental genetics CRISPR/CAS9 paired mTORC1 inhibition. To further investigate variant function in vivo, we generated a conditional Depdc5 Disease model mouse which modelled a ‘second-hit’ mechanism of disease. Generation of Depdc5 null ‘clones’ in the embryonic Mouse model brain resulted in mTORC1 hyperactivity and modelled epilepsy and FCD symptoms including large dysmorphic mTOR neurons, defective migration and lower seizure thresholds.
    [Show full text]
  • Nprl2/Tusc4 Functions As a Tumor Suppressor by Regulating Brca1's
    Texas Medical Center Library DigitalCommons@TMC UT GSBS Dissertations and Theses (Open Access) Graduate School of Biomedical Sciences 12-2014 NPRL2/TUSC4 FUNCTIONS AS A TUMOR SUPPRESSOR BY REGULATING BRCA1’S STABILITY VIA THE E3 UBIQUITINATION PATHWAY Yang Peng Follow this and additional works at: http://digitalcommons.library.tmc.edu/utgsbs_dissertations Part of the Medical Cell Biology Commons Recommended Citation Peng, Yang, "NPRL2/TUSC4 FUNCTIONS AS A TUMOR SUPPRESSOR BY REGULATING BRCA1’S STABILITY VIA THE E3 UBIQUITINATION PATHWAY" (2014). UT GSBS Dissertations and Theses (Open Access). 536. http://digitalcommons.library.tmc.edu/utgsbs_dissertations/536 This Dissertation (PhD) is brought to you for free and open access by the Graduate School of Biomedical Sciences at DigitalCommons@TMC. It has been accepted for inclusion in UT GSBS Dissertations and Theses (Open Access) by an authorized administrator of DigitalCommons@TMC. For more information, please contact [email protected]. NPRL2/TUSC4 FUNCTIONS AS A TUMOR SUPPRESSOR BY REGULATING BRCA1’S STABILITY VIA THE E3 UBIQUITINATION PATHWAY by YANG PENG, M.S. APPROVED: ______________________ Shiaw-Yih Lin, Ph.D. Supervisory Professor ______________________ Ju-Seog Lee, Ph.D. ______________________ Jessica Tyler, Ph.D. ______________________ Hui-Kuan Lin, Ph.D. ______________________ Chun Li, Ph.D. APPROVED: ______________________ Dean, The University of Texas Graduate School of Biomedical Sciences i NPRL2/TUSC4 FUNCTIONS AS A TUMOR SUPPRESSOR BY REGULATING BRCA1’S STABILITY VIA THE E3 UBIQUITINATION PATHWAY A DISSERTATION Presented to the Faculty of The University of Texas Health Science Center at Houston and The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences in Partial Fulfillment of the Requirements for the degree of DOCTOR OF PHILOSOPHY by Yang Peng, M.S.
    [Show full text]
  • Chromosome 3P and Breast Cancer
    B.J Hum Jochimsen Genet et(2002) al.: Stetteria 47:453–459 hydrogenophila © Jpn Soc Hum Genet and Springer-Verlag4600/453 2002 MINIREVIEW Qifeng Yang · Goro Yoshimura · Ichiro Mori Takeo Sakurai · Kennichi Kakudo Chromosome 3p and breast cancer Received: April 30, 2002 / Accepted: May 27, 2002 Abstract Solid tumors in humans are now believed to are now believed to develop through a multistep process develop through a multistep process that activates that activates oncogenes and inactivates tumor suppressor oncogenes and inactivates tumor suppressor genes. Loss of genes (Lopez-Otin and Diamandis 1998). Inactivation of a heterozygosity at chromosomes 3p25, 3p22–24, 3p21.3, tumor suppressor gene (TSG) often involves mutation of 3p21.2–21.3, 3p14.2, 3p14.3, and 3p12 has been reported in one allele and loss or replacement of a chromosomal seg- breast cancers. Retinoid acid receptor 2 (3p24), thyroid ment containing another allele. Loss of heterozygosity hormone receptor 1 (3p24.3), Ras association domain fam- (LOH) has been found on chromosomes 1p, 1q, 3p, 6q, 7p, ily 1A (3p21.3), and the fragile histidine triad gene (3p14.2) 11q, 13q, 16q, 17p, 17q, 18p, 18q, and 22q in breast cancer have been considered as tumor suppressor genes (TSGs) for (Smith et al. 1993; Callahan et al. 1992; Sato et al. 1990; breast cancers. Epigenetic change may play an important Hirano et al. 2001a); the commonly deleted regions include role for the inactivation of these TSGs. Screens for pro- 3p, 6q, 7p, 11q, 16q, and 17p (Smith et al. 1993; Hirano et al. moter hypermethylation may be able to identify other TSGs 2001a).
    [Show full text]
  • C-Jun Transcriptional Regulates NPRL2 to Promote the Proliferation Activity of Prostate Cancer Cells Via AKT/MDM2/P53 Signaling Pathway
    c-Jun Transcriptional Regulates NPRL2 to Promote the Proliferation Activity of Prostate Cancer Cells via AKT/MDM2/p53 Signaling Pathway Daixing Hu The First Aliated Hospital of Chongqing Medical University Li Jiang The First Aliated Hospital of Chongqing Medical University Shengjun Luo The First Aliated Hospital of Chongqing Medical University Xin Zhao The First Aliated Hospital of Chongqing Medical University Guozhi Zhao The First Aliated Hospital of Chongqing Medical University Xiaoyi Du The First Aliated Hospital of Chongqing Medical University Yu Tang The First Aliated Hospital of Chongqing Medical University Wei Tang ( [email protected] ) The First Aliated Hospital of Chongqing Medical University Research Keywords: Prostate cancer, NPRL2, Proliferation, Transcription Posted Date: November 19th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-108065/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/35 Abstract Background High NPRL2 (Nitrogen permease regulator-like 2) expression is a prognostic marker for poor clinical outcomes in prostate cancer (PCa). However, the regulatory mechanisms of NPRL2 in PCa remain unknown. Methods The expression level of NPRL2 in prostate cancer tissues were veried through The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The effect of NPRL2 gene in promoting the proliferation of prostate cancer was determined by CCK8 and clone formation assays. The apoptosis rate and cell cycle analysis were tested by flow cytometry. Luciferase reporter gene and ChIP were used to verify the binding relationship between transcription factor c-Jun and the NPRL2 5’ region. The effect of NPRL2 and the MK2206 on the AKT/MDM2/p53 signaling pathway was veried by western blotting.
    [Show full text]