In Silico Cross-Analyses with Postsynaptic Proteins and Neuropsychiatric Disorder Susceptibility Genes

Total Page:16

File Type:pdf, Size:1020Kb

In Silico Cross-Analyses with Postsynaptic Proteins and Neuropsychiatric Disorder Susceptibility Genes bioRxiv preprint doi: https://doi.org/10.1101/801563; this version posted October 11, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.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. Exploring the Pannexin 1 interactome: In silico cross-analyses with postsynaptic proteins and neuropsychiatric disorder susceptibility genes Simona D Frederiksen1, Leigh E Wicki-Stordeur1, Juan C Sanchez-Arias1, and Leigh Anne Swayne1 1Division of Medical Sciences, University of Victoria, Victoria, BC Canada The Pannexin 1 (Panx1) channel-forming protein is enriched in body, Panx1 is enriched in the central nervous system (CNS; the central nervous system, and has been associated with sev- reviewed by Boyce et al. (2018)). Within the mouse cortex, eral critical neurodevelopmental and plasticity functions; these it is highly expressed in neurons and oligodendrocytes (Ray include dendritic spine formation, neuronal network develop- et al., 2005; Vogt et al., 2005; Zhang et al., 2014; Zoidl et al., ment, synaptic plasticity, and pathological brain states such as 2007)[see brainrnaseq.org]. Consistent with its reported role ischemia, epilepsy, and neurodegeneration. Despite major ad- in regulating developmental cellular behaviours like prolif- vances in understanding the properties and activation modes eration and differentiation in a number of different cells and of Panx1, the Panx1 interactome remains largely uncharac- terized. Considering that Panx1 has been implicated in crit- tissues from other groups, work in our lab has uncovered a ical neurodevelopmental and neurodegenerative processes and role for Panx1 in regulating several aspects of neuronal de- diseases, we investigated the Panx1 interactome (482 Panx1- velopment. interacting proteins) identified from mouse N2a cells. These Our early work revealed that Panx1 promotes neural pre- proteins were cross-analyzed with the postsynaptic proteome cursor cell maintenance in vitro (Wicki-Stordeur et al., of the adult mouse brain previously identified by mass spec- 2012) and in vivo (Wicki-Stordeur et al., 2016). Con- trometry (LC-MS/MS), and neurodegenerative disease and neu- rodevelopmental disorder susceptibility genes previously iden- versely, we found that Panx1 inhibits neurite development tified by genome-wide association studies (GWAS); and then in Neuro2a (N2a) and neural precursor cells (Wicki-Stordeur further investigated using various bioinformatics tools (PAN- and Swayne, 2013). Similarly, cortical neurons from Panx1 THER, GO, KEGG and STRING databases). A total of 104 of knockout mice demonstrated an increased density of den- the Panx1-interacting proteins were located at the postsynapse, dritic spines, as well as larger groups of co-activated neu- and 99 of these formed a 16-cluster protein-protein interaction rons, also known as network ensembles (Sanchez-Arias et (PPI) network (hub proteins: Eef2, Rab6A, Ddx39b, Mapk1, al., 2019). This regulation of neurites and dendritic spines Fh1, Ndufv1 et cetera). The cross-analysis led to the discov- connects nicely with our earlier discovery of Panx1 protein- ery of proteins and candidate genes involved in synaptic func- protein interactions (PPIs) with dendritic spine regulating tion and homeostasis. Of particular note, our analyses also re- proteins, such as the actin-related protein (Arp) 2/3 com- vealed that certain Panx1-interacting proteins are implicated in plex components (Wicki-Stordeur and Swayne, 2013), and Parkinson disease, Alzheimer disease, Huntington disease, amy- otrophic lateral sclerosis, schizophrenia, autism spectrum disor- the microtubule-interacting protein collapsin-response medi- der and epilepsy. Altogether, our work revealed important clues ator protein 2 (Crmp2) (Xu et al., 2018). A role for Panx1 in to the role of Panx1 in neuronal function in health and disease by regulating neuronal development and connectivity is further expanding our knowledge of the PPI network of Panx1, and un- supported by work revealing its role in hippocampal synaptic veiling previously unidentified Panx1-interacting proteins and plasticity under naïve conditions (Ardiles et al., 2014; Ga- networks involved in biological processes and disease. jardo et al., 2018; Prochnow et al., 2012), as well as re- cently reported connections to intellectual disability (Shao Panx1 | PPIs | pathway analysis | neurodegenerative diseases | neurodevel- opmental disorders | Parkinson disease | Alzheimer disease | chaperones | et al., 2016) and autism spectrum disorder (ASD) (Davis et vesicle-mediated transport al., 2012). Moreover, Panx1 has been implicated in synap- Correspondence: Dr. Leigh Anne Swayne. Division of Medical Sciences, Uni- tic dysfunction in ischemia (MacVicar and Thompson, 2010; versity of Victoria, 3800 Finnerty Rd, Victoria BC V8P 5C2, Canada. Phone: +1 250-853-3723. E-mail: [email protected]. Twitter: @dr_swayne Weilinger et al., 2016; Weilinger et al., 2013) and has been suggested to be involved in CNS dysfunction associated with Alzheimer disease (Orellana et al., 2011b) and Parkinson dis- Introduction ease (Diaz et al., 2019). Pannexin 1 (Panx1) is a channel-forming protein that is most Given the newly-discovered role in regulating development commonly known for its role in mediating ATP release (Dahl, of dendritic spines and neuronal networks, our goal in this 2015; Lohman and Isakson, 2014) with context-specific (tis- study was to further expand our analysis of the Panx1 inter- sue and cell type expression, developmental stage and dis- actome, that we previously reported on – partially in Wicki- ease state) properties and regulation (reviewed by Chiu et Stordeur and Swayne (2013) and Wicki-Stordeur (2015), us- al. (2018)). Although ubiquitously expressed throughout the ing in silico tools (Fig. 1). Firstly, we performed over- Frederiksen et al. | bioRχiv | October 10, 2019 | 1–29 bioRxiv preprint doi: https://doi.org/10.1101/801563; this version posted October 11, 2019. 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. Fig. 1. Work flow for the current study from materials and methods to results. In this study, five subanalyses were conducted (further described in Fig. S1), which explored the: (1) Panx1 interactome in mouse N2a cells, (2) postsynaptic proteome in the adult mouse brain associated with the Panx1 interactome, and (3) neurodegenerative disease and neurodevelopmental disorder susceptibility genes associated with the Panx1 interactome. The data was obtained from three sources: Swayne lab [our lab], the genome-wide association study (GWAS) catalog (Buniello et al., 2019) and the original research article, Roy et al. (2018). The data were analyzed using the statistical computing environment R and the following databases: PANTHER [Protein ANalysis THrough Evolutionary Relationships] (Mi et al., 2019a; Mi et al., 2019b), KEGG [Kyoto Encyclopedia of Genes and Genomes] (Kanehisa et al., 2017; Kanehisa and Goto, 2000) and STRING [Search Tool for the Retrieval of Interacting Genes/Proteins] (Szklarczyk et al., 2019). In addition to the subanalyses, the findings across analyses were combined to provide a comprehensive overview for each neurodegenerative diseases and neurodevelopment disorders in relation to the Panx1 interactome. representation analyses of the entire Panx1 interactome to comparisons, the R statistical computing environment v3.6.0 get a better sense of its putative cellular and pathophysio- was applied. logical roles. Overrepresentation analyses determine if the Panx1-interacting proteins are present more than would be Identification of the Panx1 interactome in mouse N2a expected within the protein classes, cellular components and cells. We previously identified the putative Panx1 interac- biological pathways under investigation. Next, to gain fur- tome from mouse N2a neuroblastoma-derived cells (Wicki- ther insight into how Panx1 regulates dendritic spines, we Stordeur and Swayne, 2013), using methods that were com- performed a cross-analysis with the mouse brain postsynaptic prehensively described in that work as well as in Wicki- proteome, and identified links to existing PPI networks. To Stordeur (2015). Briefly, proteins co-precipitating with further examine the link between Panx1 and neurodegener- Panx1-EGFP [enhanced green fluorescent protein] or EGFP ative diseases and neurodevelopmental disorders associated from N2a cells, were identified by the UVIC-Genome with abnormalities of dendritic spines (recipients of excita- BC Proteomics Centre using high performance liquid tory inputs) (Forrest et al., 2018; Maiti et al., 2015; Pen- chromatography-tandem mass spectrometry (LC-MS/MS) zes et al., 2011), we also cross-analyzed the Panx1 interac- followed by analysis with Proteome Discoverer v1.3.0.339 tome with suggestive candidate genes identified by genome- (Thermo Scientific) and Mascot v2.2 (Perkins et al., 1999) wide association studies (GWAS). Here, we focused specifi- [percolator settings: Max delta Cn 0.05, Target false discov- cally on Parkinson disease, Alzheimer disease, amyotrophic ery rate (FDR) strict 0.01, Target FDR relaxed 0.05 with val- lateral sclerosis (ALS), Huntington disease, schizophrenia, idation based on q-value]. The q-value refers to the minimal ASD and epilepsy.
Recommended publications
  • PTPRK Expression Is Downregulated in Drug Resistant Ovarian Cancer Cell Lines, and Especially in ALDH1A1 Positive Cscs-Like Popu
    Article PTPRK Expression Is Downregulated in Drug Resistant Ovarian Cancer Cell Lines, and Especially in ALDH1A1 Positive CSCs‐Like Populations Monika Świerczewska 1,*, Karolina Sterzyńska 1, Karolina Wojtowicz 1, Dominika Kaźmierczak 1, Dariusz Iżycki 2, Michał Nowicki 1, Maciej Zabel 1,3 and Radosław Januchowski 1 1 Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61‐781 Poznań, Poland; [email protected] (K.S.); [email protected] (K.W.); [email protected] (D.K.); [email protected] (M.N.); [email protected] (M.Z.); [email protected] (R.J.) 2 Department of Cancer Immunology, Poznan University of Medical Sciences, Garbary 15 St., 61‐866 Poznań, Poland; [email protected] 3 Department of Anatomy and Histology, University of Zielona Góra, Licealna 9 St., 65‐417 Zielona Góra, Poland * Correspondence: [email protected]; Tel.: +48‐61‐8546428 Received: 26 March 2019; Accepted: 24 April 2019; Published: 25 April 2019 Abstract: Background: Ovarian cancer is the 7th most common cancer and 8th most mortal cancer among woman. The standard treatment includes cytoreduction surgery followed by chemotherapy. Unfortunately, in most cases, after treatment, cancer develops drug resistance. Decreased expression and/or activity of protein phosphatases leads to increased signal transduction and development of drug resistance in cancer cells. Methods: Using sensitive (W1, A2780) and resistant ovarian cancer cell lines, the expression of Protein Tyrosine Phosphatase Receptor Type K (PTPRK) was performed at the mRNA (real‐time PCR analysis) and protein level (Western blot, immunofluorescence analysis). The protein expression in ovarian cancer tissues was determined by immunohistochemistry.
    [Show full text]
  • Hearing Aging Is 14.1±0.4% GWAS-Heritable
    medRxiv preprint doi: https://doi.org/10.1101/2021.07.05.21260048; this version posted July 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC 4.0 International license . Predicting age from hearing test results with machine learning reveals the genetic and environmental factors underlying accelerated auditory aging Alan Le Goallec1,2+, Samuel Diai1+, Théo Vincent1, Chirag J. Patel1* 1Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA 2Department of Systems, Synthetic and Quantitative Biology, Harvard University, Cambridge, MA, 02118, USA +Co-first authors *Corresponding author Contact information: Chirag J Patel [email protected] Abstract With the aging of the world population, age-related hearing loss (presbycusis) and other hearing disorders such as tinnitus become more prevalent, leading to reduced quality of life and social isolation. Unveiling the genetic and environmental factors leading to age-related auditory disorders could suggest lifestyle and therapeutic interventions to slow auditory aging. In the following, we built the first machine learning-based hearing age predictor by training models to predict chronological age from hearing test results (root mean squared error=7.10±0.07 years; R-Squared=31.4±0.8%). We defined hearing age as the prediction outputted by the model on unseen samples, and accelerated auditory aging as the difference between a participant’s hearing age and age. We then performed a genome wide association study [GWAS] and found that accelerated hearing aging is 14.1±0.4% GWAS-heritable.
    [Show full text]
  • Biochemical and Functional Analyses of PANX1 Variants
    Western University Scholarship@Western Electronic Thesis and Dissertation Repository June 2019 Biochemical and Functional Analyses of PANX1 Variants Daniel Nouri Nejad The University of Western Ontario Supervisor Penuela, Silvia The University of Western Ontario Graduate Program in Anatomy and Cell Biology A thesis submitted in partial fulfillment of the equirr ements for the degree in Master of Science © Daniel Nouri Nejad 2019 Follow this and additional works at: https://ir.lib.uwo.ca/etd Part of the Molecular Biology Commons Recommended Citation Nouri Nejad, Daniel, "Biochemical and Functional Analyses of PANX1 Variants" (2019). Electronic Thesis and Dissertation Repository. 6230. https://ir.lib.uwo.ca/etd/6230 This Dissertation/Thesis is brought to you for free and open access by Scholarship@Western. It has been accepted for inclusion in Electronic Thesis and Dissertation Repository by an authorized administrator of Scholarship@Western. For more information, please contact [email protected]. Abstract Pannexin 1 (PANX1) is a glycoprotein capable of forming large-pore single- membrane channels permeable to signaling molecules such as ATP. In this study, we interrogated different domains by introducing naturally occurring variants reported in melanoma and assessed their impact on the channel function of PANX1 at the cell surface. From this, we discovered a novel tyrosine phosphorylation site at Tyr150, that when disrupted via a missense mutation resulted in hypo-glycosylation and a greater capacity to traffic to the cell-surface and enhanced dye uptake. We have also uncovered a highly conserved ancestral allele, Gln5His, that has a greater allele frequency than the derived allele Gln5 in global and cancer cohorts but was not associated with cancer aggressiveness.
    [Show full text]
  • A Single-Cell Transcriptional Atlas Identifies Extensive Heterogeneity in the Cellular Composition of Tendons
    bioRxiv preprint doi: https://doi.org/10.1101/801266; this version posted October 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. A single-cell transcriptional atlas identifies extensive heterogeneity in the cellular composition of tendons Jacob B Swanson1, Andrea J De Micheli2, Nathaniel P Disser1, Leandro M Martinez1, Nicholas R Walker1,3, Benjamin D Cosgrove2, Christopher L Mendias1,3,* 1Hospital for Special Surgery, New York, NY, USA 2Meining School of Biomedical Engineering, Cornell University, Ithaca, NY, USA 3Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA *Corresponding Author Christopher Mendias, PhD Hospital for Special Surgery 535 E 70th St New York, NY 10021 USA +1 212-606-1785 [email protected] Keywords: tenocyte; tendon fibroblast; pericyte; single-cell RNA sequencing bioRxiv preprint doi: https://doi.org/10.1101/801266; this version posted October 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract Tendon is a dense, hypocellular connective tissue that transmits forces between muscles and bones. Cellular heterogeneity is increasingly recognized as an important factor in the biological basis of tissue homeostasis and disease, but little is known about the diversity of cells that populate tendon. Our objective was to explore the heterogeneity of cells in mouse Achilles tendons using single-cell RNA sequencing. We identified 13 unique cell types in tendons, including 4 previously undescribed populations of fibroblasts.
    [Show full text]
  • Comprehensive Analyses of 723 Transcriptomes Enhance Genetic and Biological Interpretations for Complex Traits in Cattle
    Downloaded from genome.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press Resource Comprehensive analyses of 723 transcriptomes enhance genetic and biological interpretations for complex traits in cattle Lingzhao Fang,1,2,3,4,8 Wentao Cai,2,5,8 Shuli Liu,1,5,8 Oriol Canela-Xandri,3,4,8 Yahui Gao,1,2 Jicai Jiang,2 Konrad Rawlik,3 Bingjie Li,1 Steven G. Schroeder,1 Benjamin D. Rosen,1 Cong-jun Li,1 Tad S. Sonstegard,6 Leeson J. Alexander,7 Curtis P. Van Tassell,1 Paul M. VanRaden,1 John B. Cole,1 Ying Yu,5 Shengli Zhang,5 Albert Tenesa,3,4 Li Ma,2 and George E. Liu1 1Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, Maryland 20705, USA; 2Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland 20742, USA; 3The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian EH25 9RG, United Kingdom; 4Medical Research Council Human Genetics Unit at the Medical Research Council Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 2XU, United Kingdom; 5College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; 6Acceligen, Eagan, Minnesota 55121, USA; 7Fort Keogh Livestock and Range Research Laboratory, Agricultural Research Service, USDA, Miles City, Montana 59301, USA By uniformly analyzing 723 RNA-seq data from 91 tissues and cell types, we built a comprehensive gene atlas and studied tissue specificity of genes in cattle. We demonstrated that tissue-specific genes significantly reflected the tissue-relevant biol- ogy, showing distinct promoter methylation and evolution patterns (e.g., brain-specific genes evolve slowest, whereas testis- specific genes evolve fastest).
    [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]
  • Serum Cell-Free DNA Methylation of OPCML and HOXD9 As a Biomarker That May Aid in Differential Diagnosis Between Cholangiocarcin
    Wasenang et al. Clinical Epigenetics (2019) 11:39 https://doi.org/10.1186/s13148-019-0634-0 RESEARCH Open Access Serum cell-free DNA methylation of OPCML and HOXD9 as a biomarker that may aid in differential diagnosis between cholangiocarcinoma and other biliary diseases Wiphawan Wasenang1,2, Ponlatham Chaiyarit3, Siriporn Proungvitaya1,4 and Temduang Limpaiboon1,4* Abstract Background: Cholangiocarcinoma (CCA) is a fatal cancer of the bile duct epithelial cell lining. The misdiagnosis of CCA and other biliary diseases may occur due to the similarity of clinical manifestations and blood tests resulting in inappropriate or delayed treatment. Thus, an accurate and less-invasive method for differentiating CCA from other biliary diseases is inevitable. Methods: We quantified methylation of OPCML, HOXA9,andHOXD9 in serum cell-free DNA (cfDNA) of CCA patients and other biliary diseases using methylation-sensitive high-resolution melting (MS-HRM). Their potency as differential biomarkers between CCA and other biliary diseases was also evaluated by using receiver operating characteristic (ROC) curves. Results: The significant difference of methylation levels of OPCML and HOXD9 was observed in serum cfDNA of CCA compared to other biliary diseases. Assessment of serum cfDNA methylation of OPCML and HOXD9 as differential biomarkers of CCA and other biliary diseases showed the area under curve (AUC) of 0.850 (0.759–0.941) for OPCML which sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and accuracy were 80.00%, 90.00%, 88.88%, 81.81%, and 85.00%, respectively. The AUC of HOXD9 was 0.789 (0.686–0.892) with sensitivity, specificity, PPV, NPV, and accuracy of 67.50%, 90.00%, 87.09%, 73.46%, and 78.75%, respectively.
    [Show full text]
  • Single-Cell RNA Sequencing Demonstrates the Molecular and Cellular Reprogramming of Metastatic Lung Adenocarcinoma
    ARTICLE https://doi.org/10.1038/s41467-020-16164-1 OPEN Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma Nayoung Kim 1,2,3,13, Hong Kwan Kim4,13, Kyungjong Lee 5,13, Yourae Hong 1,6, Jong Ho Cho4, Jung Won Choi7, Jung-Il Lee7, Yeon-Lim Suh8,BoMiKu9, Hye Hyeon Eum 1,2,3, Soyean Choi 1, Yoon-La Choi6,10,11, Je-Gun Joung1, Woong-Yang Park 1,2,6, Hyun Ae Jung12, Jong-Mu Sun12, Se-Hoon Lee12, ✉ ✉ Jin Seok Ahn12, Keunchil Park12, Myung-Ju Ahn 12 & Hae-Ock Lee 1,2,3,6 1234567890():,; Advanced metastatic cancer poses utmost clinical challenges and may present molecular and cellular features distinct from an early-stage cancer. Herein, we present single-cell tran- scriptome profiling of metastatic lung adenocarcinoma, the most prevalent histological lung cancer type diagnosed at stage IV in over 40% of all cases. From 208,506 cells populating the normal tissues or early to metastatic stage cancer in 44 patients, we identify a cancer cell subtype deviating from the normal differentiation trajectory and dominating the metastatic stage. In all stages, the stromal and immune cell dynamics reveal ontological and functional changes that create a pro-tumoral and immunosuppressive microenvironment. Normal resident myeloid cell populations are gradually replaced with monocyte-derived macrophages and dendritic cells, along with T-cell exhaustion. This extensive single-cell analysis enhances our understanding of molecular and cellular dynamics in metastatic lung cancer and reveals potential diagnostic and therapeutic targets in cancer-microenvironment interactions. 1 Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea.
    [Show full text]
  • Supplementary Table 1: Adhesion Genes Data Set
    Supplementary Table 1: Adhesion genes data set PROBE Entrez Gene ID Celera Gene ID Gene_Symbol Gene_Name 160832 1 hCG201364.3 A1BG alpha-1-B glycoprotein 223658 1 hCG201364.3 A1BG alpha-1-B glycoprotein 212988 102 hCG40040.3 ADAM10 ADAM metallopeptidase domain 10 133411 4185 hCG28232.2 ADAM11 ADAM metallopeptidase domain 11 110695 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 195222 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 165344 8751 hCG20021.3 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 189065 6868 null ADAM17 ADAM metallopeptidase domain 17 (tumor necrosis factor, alpha, converting enzyme) 108119 8728 hCG15398.4 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 117763 8748 hCG20675.3 ADAM20 ADAM metallopeptidase domain 20 126448 8747 hCG1785634.2 ADAM21 ADAM metallopeptidase domain 21 208981 8747 hCG1785634.2|hCG2042897 ADAM21 ADAM metallopeptidase domain 21 180903 53616 hCG17212.4 ADAM22 ADAM metallopeptidase domain 22 177272 8745 hCG1811623.1 ADAM23 ADAM metallopeptidase domain 23 102384 10863 hCG1818505.1 ADAM28 ADAM metallopeptidase domain 28 119968 11086 hCG1786734.2 ADAM29 ADAM metallopeptidase domain 29 205542 11085 hCG1997196.1 ADAM30 ADAM metallopeptidase domain 30 148417 80332 hCG39255.4 ADAM33 ADAM metallopeptidase domain 33 140492 8756 hCG1789002.2 ADAM7 ADAM metallopeptidase domain 7 122603 101 hCG1816947.1 ADAM8 ADAM metallopeptidase domain 8 183965 8754 hCG1996391 ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) 129974 27299 hCG15447.3 ADAMDEC1 ADAM-like,
    [Show full text]
  • Co-Occurrence Based Meta-Analysis of Scientific Texts
    Vol. 21 no. 9 2005, pages 2049–2058 BIOINFORMATICS ORIGINAL PAPER doi:10.1093/bioinformatics/bti268 Data and text mining Co-occurrence based meta-analysis of scientific texts: retrieving biological relationships between genes R. Jelier1,∗ G. Jenster2, L. C. J. Dorssers3, C. C. van der Eijk1, E. M. van Mulligen1, B. Mons1 and J. A. Kors1 1Department of Medical Informatics, 2Department of Urology and 3Department of Pathology, Erasmus MC—University Medical Center, Rotterdam, The Netherlands Received on July 15, 2004; revised on December 22, 2004; accepted on January 6, 2005 Advance Access publication January 18, 2005 ABSTRACT are studied, the number of relevant publications will frequently be Motivation: The advent of high-throughput experiments in molecu- prohibitively large. This renders the traditional approach of manu- lar biology creates a need for methods to efficiently extract and use ally searching bibliographic databases for every gene and reading information for large numbers of genes. Recently, the associative scientific articles inadequate. It is therefore an important challenge concept space (ACS) has been developed for the representation of at this time to make the available information both accessible and information extracted from biomedical literature. The ACS is a Euc- interpretable for molecular biologists. lidean space in which thesaurus concepts are positioned and the An interesting current development is the use of annotations of distances between concepts indicates their relatedness. The ACS genes with gene ontology (GO) terms (Ashburner et al., 2000; Camon uses co-occurrence of concepts as a source of information. In this et al., 2004) for the analysis of the results of microarray experiments paper we evaluate how well the system can retrieve functionally (Zhang et al., 2004; Al Shahrour et al., 2004).
    [Show full text]
  • Mouse Mol.Wt.: 48 Kda Panx1 Antibody
    #M040570 Panx1 Antibody Order 021-34695924 [email protected] Support 400-6123-828 50ul [email protected] 100 uL Web www.abmart.cn Description: The pannexin gene family encodes a second class of putative gap junction proteins and are highly conserved in invertebrates and mammals. Pannexins (Panx) are four-pass transmembrane proteins that oligomerize to form large pore ion and metabolite-permeable channels. Pannexin-1 (PANX1) and Pannexin-3 are closely related, while Pannexin-2 is a more distant relation. PANX1 is a transmembrane protein that forms a mechanosensitive ATP-permeable channel between adjacent cells and in the endoplasmic reticulum. PANX1 may play a role as a Ca2+ -leak channel to regulate ER Ca2+ homeostasis and regulates neural stem and progenitor cell proliferation. Uniprot: Q96RD7 Alternative Names: innexin; MRS1; Pannexin 1; Panx1; PX1; Specificity: Antibody detects endogenous levels of total PANX1. Reactivity: Human, Mouse, Rat Source: Mouse Mol.Wt.: 48 kDa Storage Condition: Store at -20 °C. Stable for 12 months from date of receipt. 1 For in vitro research use only and not intended for use in humans or animals. Application: WB 1:1000-1:2000;IHC/IF 1:100-1:1000 Western blot analysis of PANX1 expression in SH-SY5Y whole cell lysates, 其他推荐产品 #M20001 His-Tag (2A8) Mouse mAb #M20002 Myc-Tag (19C2) Mouse mAb #M20003 HA-Tag (26D11) Mouse mAb #M20004 GFP-Tag (7G9) Mouse mAb #M20007 GST-Tag (12G8) Mouse mAb #M20008 DYDDDDDK-Tag (3B9) Mouse mAb (Binds to same epitope as Sigma’s Anti- FLAG M2 Antibody) #M20012 Anti-Myc-Tag Mouse mAb (Agarose Conjugated) #M20013 Anti-HA-Tag Mouse mAb (Agarose Conjugated) #M20018 Anti-DYKDDDDK-Tag Mouse Antibody (Agarose Conjugated) (Same as Sigma’s Anti-FLAG M2) #M20118 Anti-DYKDDDDK-Tag Mouse Antibody (Magnetic Beads) (Same as Sigma’s Anti-FLAG M2) 2 For in vitro research use only and not intended for use in humans or animals.
    [Show full text]
  • Identification of Potential Key Genes and Pathway Linked with Sporadic Creutzfeldt-Jakob Disease Based on Integrated Bioinformatics Analyses
    medRxiv preprint doi: https://doi.org/10.1101/2020.12.21.20248688; this version posted December 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Identification of potential key genes and pathway linked with sporadic Creutzfeldt-Jakob disease based on integrated bioinformatics analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice. medRxiv preprint doi: https://doi.org/10.1101/2020.12.21.20248688; this version posted December 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Abstract Sporadic Creutzfeldt-Jakob disease (sCJD) is neurodegenerative disease also called prion disease linked with poor prognosis. The aim of the current study was to illuminate the underlying molecular mechanisms of sCJD. The mRNA microarray dataset GSE124571 was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened.
    [Show full text]