Three Classes of Recurrent DNA Break Clusters in Brain Progenitors Identified by 3D Proximity-Based Break Joining Assay

Total Page:16

File Type:pdf, Size:1020Kb

Three Classes of Recurrent DNA Break Clusters in Brain Progenitors Identified by 3D Proximity-Based Break Joining Assay Three classes of recurrent DNA break clusters in brain progenitors identified by 3D proximity-based break joining assay Pei-Chi Weia,b,c,1, Cheng-Sheng Leea,b,c,1, Zhou Dua,b,c, Bjoern Schwera,b,c,2, Yuxiang Zhanga,b,c, Jennifer Kaoa,b,c, Jeffrey Zuritaa,b,c, and Frederick W. Alta,b,c,3 aHoward Hughes Medical Institute, Harvard Medical School, Boston, MA 02115; bProgram in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115; and cDepartment of Genetics, Harvard Medical School, Boston, MA 02115 Contributed by Frederick W. Alt, January 9, 2018 (sent for review November 17, 2017; reviewed by Fred H. Gage and Irving L. Weissman) We recently discovered 27 recurrent DNA double-strand break Over the past decade, we have developed and refined high- (DSB) clusters (RDCs) in mouse neural stem/progenitor cells (NSPCs). throughput genome-wide translocation sequencing (HTGTS) Most RDCs occurred across long, late-replicating RDC genes and to identify recurrent endogenous DSBs (13–15). Application of were found only after mild inhibition of DNA replication. RDC genes the HTGTS approach recently allowed us to map a set of re- share intriguing characteristics, including encoding surface proteins currently breaking genes in mouse neural stem/progenitor that organize brain architecture and neuronal junctions, and are cells (NSPCs) (16). genetically implicated in neuropsychiatric disorders and/or cancers. HTGTS maps, at nucleotide resolution, genome-wide DSBs RDC identification relies on high-throughput genome-wide trans- based on their ability to translocate to a “bait” DSB introduced location sequencing (HTGTS), which maps recurrent DSBs based on at a specific chromosomal location (13–15). Bait DSBs can be their translocation to “bait” DSBs in specific chromosomal locations. either introduced ectopically by designer nucleases (14) or pro- Cellular heterogeneity in 3D genome organization allowed unequivocal vided by endogenous DSBs, including RAG-initiated V(D)J re- identification of RDCs on 14 different chromosomes using HTGTS combination DSBs (17–19) or clusters of activation-induced baits on three mouse chromosomes. Additional candidate RDCs cytidine deaminase-initiated DSBs in IgH locus switch (S) re- were also implicated, however, suggesting that some RDCs were gions during class switch recombination (CSR) in mature B cells missed. To more completely identify RDCs, we exploited our finding (20). The ability of HTGTS to identify recurrent DSBs across the that joining of two DSBs occurs more frequently if they lie on the genome relies on cellular heterogeneity in 3D genome organization same cis chromosome. Thus, we used CRISPR/Cas9 to introduce spe- (2); however, due to the increased potential for interaction, the cific DSBs into each mouse chromosome in NSPCs that were used as joining frequency between two separate DSBs is greatly enhanced bait for HTGTS libraries. This analysis confirmed all 27 previously if the two lie on the same cis chromosome (2, 14, 17), and is en- identified RDCs and identified many new ones. NSPC RDCs fall into hanced even further if the two lie within the same topological or three groups based on length, organization, transcription level, and loop domain (2, 17, 21). Indeed, B lymphocytes exploit enhanced replication timing of genes within them. While mostly less robust, the largest group of newly defined RDCs share many intriguing Significance characteristics with the original 27. Our findings also revealed RDCs in NSPCs in the absence of induced replication stress, and support Human brain neuron genomes can differ from one another, giving the idea that the latter treatment augments an already active rise to brain mosaicism. We developed a sensitive DNA break endogenous process. joining assay that uses “bait” DNA breaks introduced on different chromosomes to detect endogenous “prey” DNA breaks across nonhomologous end-joining | neural stem cells | replication stress | the mouse brain progenitor cell genome. This approach revealed neurodevelopment | recurrent DNA break clusters 27 recurrently breaking sites, many of which occur in long neural- specific genes associated with mental illnesses and cancer. We lassical nonhomologous end-joining (C-NHEJ) is a major have exploited the finding that bait and prey DSB join more CDNA double-strand break (DSB) repair pathway in somatic frequently when on the same chromosome to increase assay cells that was first implicated based on its requisite role in V(D)J sensitivity. This approach confirms previously identified break- recombination in the developing lymphocytes (1, 2). Subse- ing neural genes and identifies new ones, often with the same quently, we found that inactivation of XRCC4, a core C-NHEJ intriguing characteristics. Our study offer potential insights into factor (3) specifically abrogates both lymphocyte and neuronal brain diversification and disease. development due to unrepaired DSBs in progenitor cells (4). Similar findings have been reported for inactivation of DNA Author contributions: P.-C.W., C.-S.L., B.S., and F.W.A. designed research; P.-C.W., C.-S.L., and J.K. performed research; P.-C.W., C.-S.L., Z.D., and J.Z. contributed new reagents/ ligase 4 (5, 6), with which XRCC4 partners in C-NHEJ end- analytic tools; P.-C.W., C.-S.L., Z.D., Y.Z., and F.W.A. analyzed data; and P.-C.W. and ligation (7). The unrepaired DSBs that cause blocked lym- F.W.A. wrote the paper. phocyte development in C-NHEJ–deficient mice are generated Reviewers: F.H.G., The Salk Institute for Biological Studies; and I.L.W., Stanford University. in antigen receptor genes by the RAG endonuclease, the pro- The authors declare no conflict of interest. tein complex that initiates V(D)J recombination (2). The na- Published under the PNAS license. NEUROSCIENCE ture of the DNA breaks that cause neuronal apoptosis in the Data deposition: Sequencing data have been deposited in the Gene Expression Omnibus absence of XRCC4 or DNA ligase 4 has remained unresolved, (GEO) database, https://www.ncbi.nlm.nih.gov/geo (accession no. GSE106822). however. Nonetheless, these previous studies promoted spec- 1P.-C.W. and C.-S.L. contributed equally to this work. ulation that specific DSBs might play a role in brain develop- 2Present address: Department of Neurological Surgery and Eli and Edythe Broad Center of ment or disease (8, 9). In this regard, more recent studies have Regeneration Medicine and Stem Cell Research, University of California, San Francisco, highlighted the potential of genomic alterations to contribute to CA 94158. brain diversification and disease (10, 11). Somatic “brain only” 3To whom correspondence should be addressed. Email: [email protected]. mutations and genomic variations also have been implicated This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. in neurodevelopmental and neuropsychiatric disorders (12). 1073/pnas.1719907115/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1719907115 PNAS | February 20, 2018 | vol. 115 | no. 8 | 1919–1924 Downloaded by guest on September 27, 2021 DSB joining within a topological domain to promote robust joining chromosome (2, 14, 17). Thus, for more complete coverage of of activation-induced cytidine deaminase-initiated S region DSBs the genome, we designed 17 additional sgRNAs to generate separated by 100 s of kb to effect exon shuffling during CSR (20), HTGTS bait DSBs, on each of the remaining 16 mouse auto- and developing lymphocytes exploit joining within chromosomal somes and the X chromosome (Fig. 1A and SI Appendix, Table loops to mediate physiological V(D)J recombination (18, 19). S1). Because the rejoining of two resected DSBs in close prox- To identify recurrent DSB clusters (RDCs) in the NSPC ge- imity to the bait break site is the most frequently detected event − − − − nome, we applied HTGTS to NSPCs from Xrcc4 / p53 / mice, in HTGTS analyses, the sgRNAs were designed to target geno- as this background enhances HTGTS detection of genomic mic sequences that were at least 5 Mb away from known RDC DSBs due to their persistence (2). HTGTS analyses from bait genes to avoid potentially confounding effects of resection events DSBs on three separate chromosomes revealed 27 RDCs found extending into adjacent RDC genes (16). In addition, to ensure by at least two of the three chromosomal baits, along with many that mapping from HTGTS bait DSBs was not influenced by additional “candidate” RDCs found with only a bait from one repetitive sequences, we selected bait genomic locales that did chromosome (16). Notably, all 27 RDCs occurred within genes not contain telomeric or simple repeat sequences. To maximize − − − − (“RDC genes”), and these genes shared an intriguing set of RDC DSB detection efficiency, we used Xrcc4 / p53 / primary characteristics, including encoding surface proteins that organize mouse NSPCs for the current HTGTS experiments, as deficiency brain architecture and neuronal junctions. Moreover, human for XRCC4 facilitates DSB persistence and detection of trans- counterparts of most mouse RDC genes had already been im- locations (2, 16). plicated genetically in neuropsychiatric disorders and cancer We used the same general approach to identify RDCs as de- (16). RDC genes also tend to be very long, moderately tran- scribed previously (16) (SI Appendix,Fig.S1). The 17 Cas9:sgRNA − − − − scribed, and late replicating. In the latter context, most RDCs constructs were introduced individually in Xrcc4 / p53 / NSPCs appeared only after
Recommended publications
  • An Integrative Prognostic and Immune Analysis of PTPRD in Pan-Cancer
    An Integrative Prognostic and Immune Analysis of PTPRD in Pan-Cancer Chunpei Ou longhua district central hospital Qin Peng longhua district central hospital Changchun Zeng ( [email protected] ) longhua district central hospital, guangdong medical university https://orcid.org/0000-0002-9489- 0627 Research Article Keywords: PTPRD, pan-cancer, prognosis, tumor-inltrating, immunotherapy Posted Date: June 4th, 2021 DOI: https://doi.org/10.21203/rs.3.rs-569409/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/22 Abstract Background: PTPRD plays an indispensable role in the occurrence of multiple tumors. However, pan- cancer analysis is unavailable. The purpose of this research was to investigate the relationship between PTPRD and immunity and describe its prognostic landscape across various tumors. Methods: We explored expression prole, survival analysis, and genomic alterations of PTPRD based on the TIMER, GEPIA, UALCAN, PrognoScan, and cBioPortal database. The frequency of PTPRD mutation and its correlation with response to immunotherapy were evaluated using the cBioPortal database. The relationship between PTPRD and immune-cell inltration was analyzed by the TIMER and TISIDB databases. A protein interaction network was constructed by the STRING database. GO and KEGG enrichment analysis was executed by the Metascape database. Results: A signicant correlation between PTPRD expression and prognosis was found in various cancers. Aberrant PRPRD expression was closely related to immune inltration. Importantly, the patients who harbored PTPRD mutation and received immune checkpoint inhibitors had worse overall survival, especially in non-small cell lung cancer and melanoma, and had a higher TMB score.
    [Show full text]
  • CCT3 Rabbit Pab
    Leader in Biomolecular Solutions for Life Science CCT3 Rabbit pAb Catalog No.: A6547 3 Publications Basic Information Background Catalog No. The protein encoded by this gene is a molecular chaperone that is a member of the A6547 chaperonin containing TCP1 complex (CCT), also known as the TCP1 ring complex (TRiC). This complex consists of two identical stacked rings, each containing eight different Observed MW proteins. Unfolded polypeptides enter the central cavity of the complex and are folded 60kDa in an ATP-dependent manner. The complex folds various proteins, including actin and tubulin. Alternate transcriptional splice variants have been characterized for this gene. Calculated MW In addition, a pseudogene of this gene has been found on chromosome 8. 56kDa/60kDa Category Primary antibody Applications WB,IHC,IF Cross-Reactivity Human, Mouse, Rat Recommended Dilutions Immunogen Information WB 1:500 - 1:2000 Gene ID Swiss Prot 7203 P49368 IHC 1:50 - 1:200 Immunogen 1:50 - 1:200 IF Recombinant fusion protein containing a sequence corresponding to amino acids 1-300 of human CCT3 (NP_005989.3). Synonyms CCT3;CCT-gamma;CCTG;PIG48;TCP-1-gamma;TRIC5 Contact Product Information www.abclonal.com Source Isotype Purification Rabbit IgG Affinity purification Storage Store at -20℃. Avoid freeze / thaw cycles. Buffer: PBS with 0.02% sodium azide,50% glycerol,pH7.3. Validation Data Western blot analysis of extracts of various cell lines, using CCT3 antibody (A6547) at 1:1000 dilution. Secondary antibody: HRP Goat Anti-Rabbit IgG (H+L) (AS014) at 1:10000 dilution. Lysates/proteins: 25ug per lane. Blocking buffer: 3% nonfat dry milk in TBST.
    [Show full text]
  • CD40 Signaling Synergizes with TLR-2 in the BCR Independent Activation of Resting B Cells
    CD40 Signaling Synergizes with TLR-2 in the BCR Independent Activation of Resting B Cells Shweta Jain, Sathi Babu Chodisetti, Javed N. Agrewala* Immunology Laboratory, Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh, India Abstract Conventionally, signaling through BCR initiates sequence of events necessary for activation and differentiation of B cells. We report an alternative approach, independent of BCR, for stimulating resting B (RB) cells, by involving TLR-2 and CD40 - molecules crucial for innate and adaptive immunity. CD40 triggering of TLR-2 stimulated RB cells significantly augments their activation, proliferation and differentiation. It also substantially ameliorates the calcium flux, antigen uptake capacity and ability of B cells to activate T cells. The survival of RB cells was improved and it increases the number of cells expressing activation induced deaminase (AID), signifying class switch recombination (CSR). Further, we also observed increased activation rate and decreased threshold period required for optimum stimulation of RB cells. These results corroborate well with microarray gene expression data. This study provides novel insights into coordination between the molecules of innate and adaptive immunity in activating B cells, in a BCR independent manner. This strategy can be exploited to design vaccines to bolster B cell activation and antigen presenting efficiency, leading to faster and better immune response. Citation: Jain S, Chodisetti SB, Agrewala JN (2011) CD40 Signaling Synergizes with TLR-2 in the BCR Independent Activation of Resting B Cells. PLoS ONE 6(6): e20651. doi:10.1371/journal.pone.0020651 Editor: Leonardo A. Sechi, Universita di Sassari, Italy Received April 14, 2011; Accepted May 6, 2011; Published June 2, 2011 Copyright: ß 2011 Jain et al.
    [Show full text]
  • An Overview of the Role of Hdacs in Cancer Immunotherapy
    International Journal of Molecular Sciences Review Immunoepigenetics Combination Therapies: An Overview of the Role of HDACs in Cancer Immunotherapy Debarati Banik, Sara Moufarrij and Alejandro Villagra * Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, 800 22nd St NW, Suite 8880, Washington, DC 20052, USA; [email protected] (D.B.); [email protected] (S.M.) * Correspondence: [email protected]; Tel.: +(202)-994-9547 Received: 22 March 2019; Accepted: 28 April 2019; Published: 7 May 2019 Abstract: Long-standing efforts to identify the multifaceted roles of histone deacetylase inhibitors (HDACis) have positioned these agents as promising drug candidates in combatting cancer, autoimmune, neurodegenerative, and infectious diseases. The same has also encouraged the evaluation of multiple HDACi candidates in preclinical studies in cancer and other diseases as well as the FDA-approval towards clinical use for specific agents. In this review, we have discussed how the efficacy of immunotherapy can be leveraged by combining it with HDACis. We have also included a brief overview of the classification of HDACis as well as their various roles in physiological and pathophysiological scenarios to target key cellular processes promoting the initiation, establishment, and progression of cancer. Given the critical role of the tumor microenvironment (TME) towards the outcome of anticancer therapies, we have also discussed the effect of HDACis on different components of the TME. We then have gradually progressed into examples of specific pan-HDACis, class I HDACi, and selective HDACis that either have been incorporated into clinical trials or show promising preclinical effects for future consideration.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Supp Table 1.Pdf
    Upregulated genes in Hdac8 null cranial neural crest cells fold change Gene Symbol Gene Title 134.39 Stmn4 stathmin-like 4 46.05 Lhx1 LIM homeobox protein 1 31.45 Lect2 leukocyte cell-derived chemotaxin 2 31.09 Zfp108 zinc finger protein 108 27.74 0710007G10Rik RIKEN cDNA 0710007G10 gene 26.31 1700019O17Rik RIKEN cDNA 1700019O17 gene 25.72 Cyb561 Cytochrome b-561 25.35 Tsc22d1 TSC22 domain family, member 1 25.27 4921513I08Rik RIKEN cDNA 4921513I08 gene 24.58 Ofa oncofetal antigen 24.47 B230112I24Rik RIKEN cDNA B230112I24 gene 23.86 Uty ubiquitously transcribed tetratricopeptide repeat gene, Y chromosome 22.84 D8Ertd268e DNA segment, Chr 8, ERATO Doi 268, expressed 19.78 Dag1 Dystroglycan 1 19.74 Pkn1 protein kinase N1 18.64 Cts8 cathepsin 8 18.23 1500012D20Rik RIKEN cDNA 1500012D20 gene 18.09 Slc43a2 solute carrier family 43, member 2 17.17 Pcm1 Pericentriolar material 1 17.17 Prg2 proteoglycan 2, bone marrow 17.11 LOC671579 hypothetical protein LOC671579 17.11 Slco1a5 solute carrier organic anion transporter family, member 1a5 17.02 Fbxl7 F-box and leucine-rich repeat protein 7 17.02 Kcns2 K+ voltage-gated channel, subfamily S, 2 16.93 AW493845 Expressed sequence AW493845 16.12 1600014K23Rik RIKEN cDNA 1600014K23 gene 15.71 Cst8 cystatin 8 (cystatin-related epididymal spermatogenic) 15.68 4922502D21Rik RIKEN cDNA 4922502D21 gene 15.32 2810011L19Rik RIKEN cDNA 2810011L19 gene 15.08 Btbd9 BTB (POZ) domain containing 9 14.77 Hoxa11os homeo box A11, opposite strand transcript 14.74 Obp1a odorant binding protein Ia 14.72 ORF28 open reading
    [Show full text]
  • 1 Supporting Information for a Microrna Network Regulates
    Supporting Information for A microRNA Network Regulates Expression and Biosynthesis of CFTR and CFTR-ΔF508 Shyam Ramachandrana,b, Philip H. Karpc, Peng Jiangc, Lynda S. Ostedgaardc, Amy E. Walza, John T. Fishere, Shaf Keshavjeeh, Kim A. Lennoxi, Ashley M. Jacobii, Scott D. Rosei, Mark A. Behlkei, Michael J. Welshb,c,d,g, Yi Xingb,c,f, Paul B. McCray Jr.a,b,c Author Affiliations: Department of Pediatricsa, Interdisciplinary Program in Geneticsb, Departments of Internal Medicinec, Molecular Physiology and Biophysicsd, Anatomy and Cell Biologye, Biomedical Engineeringf, Howard Hughes Medical Instituteg, Carver College of Medicine, University of Iowa, Iowa City, IA-52242 Division of Thoracic Surgeryh, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada-M5G 2C4 Integrated DNA Technologiesi, Coralville, IA-52241 To whom correspondence should be addressed: Email: [email protected] (M.J.W.); yi- [email protected] (Y.X.); Email: [email protected] (P.B.M.) This PDF file includes: Materials and Methods References Fig. S1. miR-138 regulates SIN3A in a dose-dependent and site-specific manner. Fig. S2. miR-138 regulates endogenous SIN3A protein expression. Fig. S3. miR-138 regulates endogenous CFTR protein expression in Calu-3 cells. Fig. S4. miR-138 regulates endogenous CFTR protein expression in primary human airway epithelia. Fig. S5. miR-138 regulates CFTR expression in HeLa cells. Fig. S6. miR-138 regulates CFTR expression in HEK293T cells. Fig. S7. HeLa cells exhibit CFTR channel activity. Fig. S8. miR-138 improves CFTR processing. Fig. S9. miR-138 improves CFTR-ΔF508 processing. Fig. S10. SIN3A inhibition yields partial rescue of Cl- transport in CF epithelia.
    [Show full text]
  • Anti-VAV3 (Aa 567-578) Polyclonal Antibody (DPABH-12442) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Anti-VAV3 (aa 567-578) polyclonal antibody (DPABH-12442) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Antigen Description Exchange factor for GTP-binding proteins RhoA, RhoG and, to a lesser extent, Rac1. Binds physically to the nucleotide-free states of those GTPases. Plays an important role in angiogenesis. Its recruitement by phosphorylated EPHA2 is critical for EFNA1-induced RAC1 GTPase activation and vascular endothelial cell migration and assembly. Immunogen Synthetic peptide: CSGEQGTLKLPEK, corresponding to internal sequence amino acids 567-578 of Human VAV3 Isotype IgG Source/Host Goat Species Reactivity Human Purification Immunogen affinity purified Conjugate Unconjugated Applications WB, IHC-P Format Liquid Size 50 μg Buffer pH: 7.40; Constituents: 0.5% BSA, Tris buffered saline Preservative 0.02% Sodium Azide Storage Store at 4°C or at -20°C for long term storage. GENE INFORMATION Gene Name VAV3 vav 3 guanine nucleotide exchange factor [ Homo sapiens ] Official Symbol VAV3 Synonyms VAV3; vav 3 guanine nucleotide exchange factor; vav 3 oncogene; guanine nucleotide exchange factor VAV3; VAV-3; FLJ40431; 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 1 © Creative Diagnostics All Rights Reserved Entrez Gene ID 10451 Protein Refseq NP_001073343 UniProt ID Q9UKW4 Chromosome Location 1p13.3 Pathway B cell receptor signaling pathway; Cell death signalling via NRAGE, NRIF and NADE; Chemokine signaling pathway; Coregulation of Androgen receptor activity; EGFR1 Signaling Pathway; Function GTPase activator activity; Rac guanyl-nucleotide exchange factor activity; SH3/SH2 adaptor activity; epidermal growth factor receptor binding; guanyl-nucleotide exchange factor activity; metal ion binding; protein binding 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 2 © Creative Diagnostics All Rights Reserved.
    [Show full text]
  • The Metabolic Regulator Histone Deacetylase 9 Contributes to Glucose Homeostasis
    Page 1 of 53 Diabetes The metabolic regulator histone deacetylase 9 contributes to glucose homeostasis abnormality induced by hepatitis C virus infection Jizheng CHEN2, Ning WANG1, Mei DONG1, Min GUO2, Yang ZHAO3, Zhiyong ZHUO3, Chao ZHANG3, Xiumei CHI4, Yu PAN4, Jing JIANG4, Hong TANG2, Junqi NIU4, Dongliang YANG5, Zhong LI1, Xiao HAN1, Qian WANG1* and Xinwen Chen2 1 Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 210029, China 2 State Key Lab of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China 3 Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China 4 Department of Hepatology, The First Hospital of Jilin University, Changchun, 130021, China 5 Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China 1 Diabetes Publish Ahead of Print, published online September 29, 2015 Diabetes Page 2 of 53 *Correspondence: Qian WANG, Ph.D. Jiangsu Province Key Lab of Human Functional Genomics Department of Biochemistry and Molecular Biology Nanjing Medical University Nanjing 210029, China E-mail: [email protected] Fax: +86-25-8362 1065 Tel: +86-25-8686 2729 2 Page 3 of 53 Diabetes ABSTRACT Class IIa histone deacetylases (HDACs), such as HDAC4, HDAC5, and HDAC7 provide critical mechanisms for regulating glucose homeostasis. Here we report HDAC9, another class IIa HDAC, regulates hepatic gluconeogenesis via deacetylation of a Forkhead box O (FoxO) family transcription factor, FoxO1, together with HDAC3. Specifically, HDAC9 expression can be strongly induced upon hepatitis C virus (HCV) infection.
    [Show full text]
  • PTEN, and KCTD13 and RAF1) That Significantly Enhanced Or Suppressed Cell Proliferation Phenotypes
    bioRxiv preprint doi: https://doi.org/10.1101/185355; this version posted September 20, 2017. 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-NC-ND 4.0 International license. Pervasive epistasis modulates neurodevelopmental defects of the autism-associated 16p11.2 deletion Janani Iyer1,9, Mayanglambam Dhruba Singh1,9, Matthew Jensen1,2,9, Payal Patel1,9, Lucilla Pizzo1, Emily Huber1, Haley Koerselman3, Alexis T. Weiner1, Paola Lepanto4, Komal Vadodaria1, Alexis Kubina1, Qingyu Wang1,2, Abigail Talbert1, Sneha Yennawar1, Jose Badano4, J. Robert Manak3,5, Melissa M. Rolls1, Arjun Krishnan6,7, and Santhosh Girirajan1,2,8* 1. Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802 2. Bioinformatics and Genomics Program, Huck Institute of Life Sciences, The Pennsylvania State University, University Park, PA 16802 3. Departments of Biology, University of Iowa, Iowa City, IA 52242 4. Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay 5. Department of Pediatrics, University of Iowa, Iowa City, IA 52242 6. Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824 7. Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 8. Department of Anthropology, The Pennsylvania State University, University Park, PA 16802 9. These authors contributed equally to this work. Correspondence to: Santhosh Girirajan 205A Life Sciences Building The Pennsylvania State University University Park, PA 16802 E-mail: [email protected] Phone: 814-865-0674 1 bioRxiv preprint doi: https://doi.org/10.1101/185355; this version posted September 20, 2017.
    [Show full text]
  • The Regulatory Roles of Phosphatases in Cancer
    Oncogene (2014) 33, 939–953 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc REVIEW The regulatory roles of phosphatases in cancer J Stebbing1, LC Lit1, H Zhang, RS Darrington, O Melaiu, B Rudraraju and G Giamas The relevance of potentially reversible post-translational modifications required for controlling cellular processes in cancer is one of the most thriving arenas of cellular and molecular biology. Any alteration in the balanced equilibrium between kinases and phosphatases may result in development and progression of various diseases, including different types of cancer, though phosphatases are relatively under-studied. Loss of phosphatases such as PTEN (phosphatase and tensin homologue deleted on chromosome 10), a known tumour suppressor, across tumour types lends credence to the development of phosphatidylinositol 3--kinase inhibitors alongside the use of phosphatase expression as a biomarker, though phase 3 trial data are lacking. In this review, we give an updated report on phosphatase dysregulation linked to organ-specific malignancies. Oncogene (2014) 33, 939–953; doi:10.1038/onc.2013.80; published online 18 March 2013 Keywords: cancer; phosphatases; solid tumours GASTROINTESTINAL MALIGNANCIES abs in sera were significantly associated with poor survival in Oesophageal cancer advanced ESCC, suggesting that they may have a clinical utility in Loss of PTEN (phosphatase and tensin homologue deleted on ESCC screening and diagnosis.5 chromosome 10) expression in oesophageal cancer is frequent, Cao et al.6 investigated the role of protein tyrosine phosphatase, among other gene alterations characterizing this disease. Zhou non-receptor type 12 (PTPN12) in ESCC and showed that PTPN12 et al.1 found that overexpression of PTEN suppresses growth and protein expression is higher in normal para-cancerous tissues than induces apoptosis in oesophageal cancer cell lines, through in 20 ESCC tissues.
    [Show full text]
  • PSD-95 Binding Dynamically Regulates NLGN1 Trafficking and Function
    PSD-95 binding dynamically regulates NLGN1 trafficking and function Jaehoon Jeonga, Saurabh Pandeya, Yan Lia, John D. Badger IIa, Wei Lua, and Katherine W. Rochea,1 aNational Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892 Edited by Solomon H. Snyder, Johns Hopkins University School of Medicine, Baltimore, MD, and approved May 3, 2019 (received for review December 20, 2018) PSD-95 is a scaffolding protein that regulates the synaptic localization necessary for maintaining presynaptic release probability (15). of many receptors, channels, and signaling proteins. The NLGN gene Meanwhile, synaptic targeting of NLGN1 is known to be in- family encodes single-pass transmembrane postsynaptic cell adhesion dependent of PSD-95, as PSD-95 is recruited to the plasma molecules that are important for synapse assembly and function. At membrane after NLGN1 (13, 16, 17). In addition, non-PDZ excitatory synapses, NLGN1 mediates transsynaptic binding with ligand-dependent NLGN1 and NLGN3 functions have been in- neurexin, a presynaptic cell adhesion molecule, and also binds to vestigated (18), suggesting a complicated physiological interplay PSD-95, although the relevance of the PSD-95 interaction is not clear. between NLGNs and PSD-95. We now show that disruption of the NLGN1 and PSD-95 interaction Posttranslational modifications have been reported for several decreases surface expression of NLGN1 in cultured neurons. Further- NLGN isoforms and are postulated to confer distinct properties (11, more, PKA phosphorylates NLGN1 on S839, near the PDZ ligand, and 12). For example, phosphorylation of the cytoplasmic region of dynamically regulates PSD-95 binding. A phosphomimetic mutation NLGN1 has recently emerged as a mechanism for isoform-specific of NLGN1 S839 significantly reduced PSD-95 binding.
    [Show full text]