Multidimensional Genetic Analysis of Repeated Seizures in the Hybrid Mouse Diversity Panel 10 Reveals a Novel Epileptogenesis Susceptibility Locus

Total Page:16

File Type:pdf, Size:1020Kb

Multidimensional Genetic Analysis of Repeated Seizures in the Hybrid Mouse Diversity Panel 10 Reveals a Novel Epileptogenesis Susceptibility Locus G3: Genes|Genomes|Genetics Early Online, published on June 15, 2017 as doi:10.1534/g3.117.042234 1 2 3 4 5 6 7 8 9 Multidimensional genetic analysis of repeated seizures in the hybrid mouse diversity panel 10 reveals a novel epileptogenesis susceptibility locus. 11 Russell J Ferland*, †, ‡, Jason Smith§, Dominick Papandrea‡, Jessica Gracias††, Leah 12 Hains§, Sridhar B Kadiyala*, Brittany O’Brien††, Eun Yong Kang‡‡, Barbara S Beyer*, and Bruce 13 J Herron§,†† 14 15 * Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 16 Albany, New York 12208 17 † Department of Neurology, Albany Medical College, Albany, New York 12208 18 ‡ Department of Biological Sciences, Center for Biotechnology and Interdisciplinary 19 Studies, Rensselaer Polytechnic Institute, Troy, New York 12180 20 § Wadsworth Center, Albany, New York 12201 21 †† Department of Biomedical Sciences, School of Public Health, SUNY Albany, Albany, New 22 York 12201 23 ‡‡ Department of Computer Science, University of California, Los Angeles, CA, USA 24 90095 25 © The Author(s) 2013. Published by the Genetics Society of America. 26 27 28 Short running title: Camta1 modifies epileptogenesis 29 Keywords: Genetics 30 Epilepsy 31 Preclinical model 32 Neuronal plasticity 33 Complex traits 34 35 36 Correspondence to either: 37 38 Russell J. Ferland, PhD Bruce J. Herron, PhD 39 Albany Medical College Center for Medical Sciences 40 Department of Neuroscience and Wadsworth Center, NYS Dept. of Health 41 Experimental Therapeutics 150 New Scotland Ave 42 47 New Scotland Avenue, MC-136 Albany, NY 12208 43 Albany, NY 12208 [email protected] 44 [email protected] 45 46 47 48 49 50 Conflict of Interest: The authors declare no competing financial interests. 51 52 Acknowledgements: This work was supported by an NIH/NINDS R01NS064283 grant to RJF. 53 We are grateful to Veterinary Sciences at the Wadsworth Center and Albany Medical College 54 that supported the completion of this work. We also thank Dr. Rob Williams for helpful 55 discussion during the progress of this study. The exceptional animal husbandry expertise 56 of Barbara Beyer, who has since passed away, was critical to the success of this 57 project. 2 58 Abstract 59 Epilepsy has many causes and comorbidities affecting as many as 4% of people in their lifetime. 60 Both idiopathic and symptomatic epilepsies are highly heritable, but genetic factors are difficult 61 to characterize among humans due to complex disease etiologies. Rodent genetic studies have 62 been critical to the discovery of seizure susceptibility loci, including Kcnj10 mutations identified 63 in both mouse and human cohorts. However, genetic analyses of epilepsy phenotypes in mice 64 to date have been carried out as acute studies in seizure-naive animals or in Mendelian models 65 of epilepsy, while humans with epilepsy have a history of recurrent seizures that also modify 66 brain physiology. We have applied a repeated seizure model to a genetic reference population, 67 following seizure susceptibility over a 36-day period. Initial differences in generalized seizure 68 threshold among the Hybrid Mouse Diversity Panel were associated with a well-characterized 69 seizure susceptibility locus found in mice; Seizure susceptibility 1. Remarkably, Szs1 influence 70 diminished as subsequent induced seizures had diminishing latencies in certain HMDP strains. 71 Administration of eight seizures, followed by an incubation period and an induced retest seizure, 72 revealed novel associations within the calmodulin-activated transcription factor 1, Camta1. Using 73 systems genetics, we have identified 4 candidate genes that are differentially expressed 74 between seizure-sensitive and -resistant strains close to Esf1 that may act individually or as a 75 coordinated response to the neuronal stress of seizures. 3 76 Significance Statement 77 Epileptogenesis is a complex trait with a partially characterized genetic basis. While recurrent 78 seizures define epilepsy, no preclinical epilepsy models have investigated if repeated seizures 79 contribute to disease progression. We used genome-wide association studies in mice to uncover 80 genetic factors that mediate acute seizure susceptibility and those dependent on repeated 81 seizures. We found that seizure-naive animals have seizure susceptibilities consistent with other 82 experimental epilepsy models. However, inducing repeated seizures shifts genetic associations 83 to a novel genomic region containing genes suitable for facilitating epileptogenesis. We show 84 that initial seizures likely contribute to epileptogenesis susceptibility and have utilized systems- 85 genetics to identify epileptogenesis candidate genes. 4 86 Introduction 87 Epilepsies in humans are highly heritable syndromes that are modified by complex 88 interactions between genes and the environment. In the US, approximately 2.2 million people 89 will develop some form of epilepsy during their lifetime (England et al. 2012). Despite diverse 90 treatments options, largely focused on seizure suppression, individuals with epilepsy suffer 91 multiple quality of life issues, refractory seizures, comorbid complications, and a higher incidence 92 of sudden death (Gupta et al. 2016). Understanding interactions between genetic seizure 93 susceptibility loci and environmental stressors induced by repeated seizures will uncover novel 94 mechanisms of epilepsy pathogenesis. This information will improve therapeutic options by 95 focusing treatment on epileptogenesis (Ferraro 2015). 96 Identification of gene by environment interactions for individuals with epilepsy is limited by 97 complex disease etiologies, and the inability to identify at risk populations prior to disease onset. 98 Consequently, the majority of epilepsy genome wide association studies (GWAS) carried out to 99 date compare affected populations to seizure-naive controls rather than following epilepsy 100 progression in a population from a seizure-naive to an epileptic state (International League 101 Against Epilepsy Consortium on Complex Epilepsies. 2014). Experimental seizure paradigms 102 can overcome such limitations to discover the genetic basis of epileptogenesis. 103 Preclinical epilepsy models have implicated multiple gene mutations as contributors to 104 seizure disorders. This success is due in part to strong genetic influences that segregate among 105 rodent models of seizure susceptibility (Engstrom and Woodbury 1988; Kosobud and Crabbe 106 1990). Unfortunately, most of these models utilized either acute seizure measurements or status 107 epilepticus as quantitative measures of seizure susceptibility (Grone and Baraban 2015). 108 Alternatively, animal models that measure epileptogenesis and other models of brain plasticity 109 are typically designed to minimize genetic background effects (Grone and Baraban 2015). 5 110 Notably, while numerous hyperexcitability factors are now known, genetic factors influencing 111 epileptogenesis remain largely uncharacterized. 112 We have utilized a repeated flurothyl-induced seizure model delivered to the Hybrid Mouse 113 Diversity Panel (HMDP) to identify genetic susceptibility factors that modify epileptogenesis 114 (Papandrea et al. 2009). Genetic differences in initial flurothyl-induced seizures were consistent 115 with previous studies showing the strongest quantitative trait loci (QTL) mapping to the Szs1 116 locus (Ferraro et al. 1997, 2004; Chaix et al. 2007). Surprisingly, as additional seizures were 117 administered to this population, the effects of Szs1 diminished, and genetic associations on 118 distal chromosome 4 garnered influence over seizure susceptibility. We attribute the shift in 119 susceptibility to seizure-related changes in brain plasticity that are modified by gene(s) that may 120 mitigate the effects of Szs1. This hypothesis is supported by interactions between Szs1 and our 121 novel epileptogenesis susceptibility locus on chromosome 4; Epileptogenesis susceptibility 122 factor 1 (Esf1). Systems genetics analysis of the Esf1 region has uncovered a co-regulated set 123 of expression QTL (eQTL) that have been implicated in neurological stress response that are 124 now also implicated in epileptogenesis. 125 126 Materials and Methods 127 Animals. The rationale for using the HMDP and its advantages with respect to statistical power 128 and genetic resolution have been previously described (Bennett et al. 2010). The inbred strains 129 of mice that were used for this study include the following: Balb/cJ (Balb; n = 8); BTBR T+Ltpr3tf/J 130 (BTBR; n = 6); BXD#/TyJ (BXD#; 30 strains, average n/strain ≤ 6 ); BXD##/RwwJ (BXD##; 18 131 strains, average n/strain ≤ 4); C57BL/6J (B6; n = 10); C57BL/10SNJ (10SNJ; n = 12); C57BL/10J 132 (10J; n = 8); C57BL/6NJ (6NJ; n= 16); C57BLKS/J (KS/J; n = 15); CAST/EiJ (CAST; n = 6); 133 CBA/J (CBA; n = 17); CE/J (CE; n = 11); DBA/2J (D2; n = 14); FVB/NJ (FVB; n = 12); LG/J (LG; 6 134 n = 19); MRL/MpJ (MRL; n = 6); NOD/ShiLtJ (NOD; n = 13); NZW/LacJ (NZW; n = 10); PWD/PhJ 135 (PWD; n = 9) and Sm/J (Sm; n = 13). Male and female mice were obtained from the Jackson 136 Laboratories (Bar Harbor, ME, USA) and 1) were acclimated to the animal facility for one week 137 before seizure testing commenced, or 2) were further bred at the Wadsworth Center and 138 exposed to the repeated-flurothyl seizure model. Mice were housed on a 12 hr light-dark cycle 139 with ad libitum access to food and water. 140 A [B6XD2]F2 cross was generated by intercrossing [B6XD2]F1 parents. Both male
Recommended publications
  • The Mineralocorticoid Receptor Leads to Increased Expression of EGFR
    www.nature.com/scientificreports OPEN The mineralocorticoid receptor leads to increased expression of EGFR and T‑type calcium channels that support HL‑1 cell hypertrophy Katharina Stroedecke1,2, Sandra Meinel1,2, Fritz Markwardt1, Udo Kloeckner1, Nicole Straetz1, Katja Quarch1, Barbara Schreier1, Michael Kopf1, Michael Gekle1 & Claudia Grossmann1* The EGF receptor (EGFR) has been extensively studied in tumor biology and recently a role in cardiovascular pathophysiology was suggested. The mineralocorticoid receptor (MR) is an important efector of the renin–angiotensin–aldosterone‑system and elicits pathophysiological efects in the cardiovascular system; however, the underlying molecular mechanisms are unclear. Our aim was to investigate the importance of EGFR for MR‑mediated cardiovascular pathophysiology because MR is known to induce EGFR expression. We identifed a SNP within the EGFR promoter that modulates MR‑induced EGFR expression. In RNA‑sequencing and qPCR experiments in heart tissue of EGFR KO and WT mice, changes in EGFR abundance led to diferential expression of cardiac ion channels, especially of the T‑type calcium channel CACNA1H. Accordingly, CACNA1H expression was increased in WT mice after in vivo MR activation by aldosterone but not in respective EGFR KO mice. Aldosterone‑ and EGF‑responsiveness of CACNA1H expression was confrmed in HL‑1 cells by Western blot and by measuring peak current density of T‑type calcium channels. Aldosterone‑induced CACNA1H protein expression could be abrogated by the EGFR inhibitor AG1478. Furthermore, inhibition of T‑type calcium channels with mibefradil or ML218 reduced diameter, volume and BNP levels in HL‑1 cells. In conclusion the MR regulates EGFR and CACNA1H expression, which has an efect on HL‑1 cell diameter, and the extent of this regulation seems to depend on the SNP‑216 (G/T) genotype.
    [Show full text]
  • Pharmacogenomics of Drug-Induced Conditions
    Pharmacogenomics of Drug-Induced Conditions Dan M. Roden, M.D. Professor of Medicine and Pharmacology Director, Oates Institute for Experimental Therapeutics Assistant Vice-Chancellor for Personalized Medicine Vanderbilt University School of Medicine !1 An “idiosyncratic” drug response AR, 78 year old male • Coronary artery disease; 13 years s/p bypass surgery • 2 days after starting dofetilide (potent IKr blocker)… No personal or family history of syncope, sudden death • KCNQ1 variant leading to R583C identified • In vitro: ↓IKs • Not found in >400 ethnically-matched controls • ∴this is likely subclinical congenital Long QT Syndrome !2 Another face of the congenital long QT syndrome !3 Father’s ECG !4 Mother’s ECG !5 9-year-old sister’s ECG !6 Family tree Aunt, cousin, niece with 402 426 syncope 462 405 439 428 (RBBB) 436 428 492 411 419 404 • One episode of syncope, age 20 • Multiple ER visits for acute allergic reactions à epinephrine without incident !7 Mother’s ECG post-exercise !8 Identifying a mutation in KCNQ1 mothersister …YI G FLGLI… …YI D FLGLI… G269D !9 Family tree Aunt, cousin, niece with 402 426 syncope 462 405 439 428 (RBBB) 436 428 492 411 419 404 Incomplete penetrance !10 Grandfather’s ECG !11 Defining the “drug-induced long QT syndrome” • Marked QT prolongation and typical pause-dependent torsades de pointes with drug challenge, both resolving with drug withdrawal • Variants: • Marked QT prolongation (e.g. >520 or 550 or 600 msec), recognized and drug withdrawn (no torsades) • pause-dependent polymorphic VT after administration
    [Show full text]
  • Aquaporin Channels in the Heart—Physiology and Pathophysiology
    International Journal of Molecular Sciences Review Aquaporin Channels in the Heart—Physiology and Pathophysiology Arie O. Verkerk 1,2,* , Elisabeth M. Lodder 2 and Ronald Wilders 1 1 Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; [email protected] 2 Department of Experimental Cardiology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; [email protected] * Correspondence: [email protected]; Tel.: +31-20-5664670 Received: 29 March 2019; Accepted: 23 April 2019; Published: 25 April 2019 Abstract: Mammalian aquaporins (AQPs) are transmembrane channels expressed in a large variety of cells and tissues throughout the body. They are known as water channels, but they also facilitate the transport of small solutes, gasses, and monovalent cations. To date, 13 different AQPs, encoded by the genes AQP0–AQP12, have been identified in mammals, which regulate various important biological functions in kidney, brain, lung, digestive system, eye, and skin. Consequently, dysfunction of AQPs is involved in a wide variety of disorders. AQPs are also present in the heart, even with a specific distribution pattern in cardiomyocytes, but whether their presence is essential for proper (electro)physiological cardiac function has not intensively been studied. This review summarizes recent findings and highlights the involvement of AQPs in normal and pathological cardiac function. We conclude that AQPs are at least implicated in proper cardiac water homeostasis and energy balance as well as heart failure and arsenic cardiotoxicity. However, this review also demonstrates that many effects of cardiac AQPs, especially on excitation-contraction coupling processes, are virtually unexplored.
    [Show full text]
  • Transcriptomic Analysis of the Aquaporin (AQP) Gene Family
    Pancreatology 19 (2019) 436e442 Contents lists available at ScienceDirect Pancreatology journal homepage: www.elsevier.com/locate/pan Transcriptomic analysis of the Aquaporin (AQP) gene family interactome identifies a molecular panel of four prognostic markers in patients with pancreatic ductal adenocarcinoma Dimitrios E. Magouliotis a, b, Vasiliki S. Tasiopoulou c, Konstantinos Dimas d, * Nikos Sakellaridis d, Konstantina A. Svokos e, Alexis A. Svokos f, Dimitris Zacharoulis b, a Division of Surgery and Interventional Science, Faculty of Medical Sciences, UCL, London, UK b Department of Surgery, University of Thessaly, Biopolis, Larissa, Greece c Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece d Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece e The Warren Alpert Medical School of Brown University, Providence, RI, USA f Riverside Regional Medical Center, Newport News, VA, USA article info abstract Article history: Background: This study aimed to assess the differential gene expression of aquaporin (AQP) gene family Received 14 October 2018 interactome in pancreatic ductal adenocarcinoma (PDAC) using data mining techniques to identify novel Received in revised form candidate genes intervening in the pathogenicity of PDAC. 29 January 2019 Method: Transcriptome data mining techniques were used in order to construct the interactome of the Accepted 9 February 2019 AQP gene family and to determine which genes members are differentially expressed in PDAC as Available online 11 February 2019 compared to controls. The same techniques were used in order to evaluate the potential prognostic role of the differentially expressed genes. Keywords: PDAC Results: Transcriptome microarray data of four GEO datasets were incorporated, including 142 primary Aquaporin tumor samples and 104 normal pancreatic tissue samples.
    [Show full text]
  • Supplementary Materials
    1 Supplementary Materials: Supplemental Figure 1. Gene expression profiles of kidneys in the Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice. (A) A heat map of microarray data show the genes that significantly changed up to 2 fold compared between Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice (N=4 mice per group; p<0.05). Data show in log2 (sample/wild-type). 2 Supplemental Figure 2. Sting signaling is essential for immuno-phenotypes of the Fcgr2b-/-lupus mice. (A-C) Flow cytometry analysis of splenocytes isolated from wild-type, Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice at the age of 6-7 months (N= 13-14 per group). Data shown in the percentage of (A) CD4+ ICOS+ cells, (B) B220+ I-Ab+ cells and (C) CD138+ cells. Data show as mean ± SEM (*p < 0.05, **p<0.01 and ***p<0.001). 3 Supplemental Figure 3. Phenotypes of Sting activated dendritic cells. (A) Representative of western blot analysis from immunoprecipitation with Sting of Fcgr2b-/- mice (N= 4). The band was shown in STING protein of activated BMDC with DMXAA at 0, 3 and 6 hr. and phosphorylation of STING at Ser357. (B) Mass spectra of phosphorylation of STING at Ser357 of activated BMDC from Fcgr2b-/- mice after stimulated with DMXAA for 3 hour and followed by immunoprecipitation with STING. (C) Sting-activated BMDC were co-cultured with LYN inhibitor PP2 and analyzed by flow cytometry, which showed the mean fluorescence intensity (MFI) of IAb expressing DC (N = 3 mice per group). 4 Supplemental Table 1. Lists of up and down of regulated proteins Accession No.
    [Show full text]
  • Expression Profiling of Ion Channel Genes Predicts Clinical Outcome in Breast Cancer
    UCSF UC San Francisco Previously Published Works Title Expression profiling of ion channel genes predicts clinical outcome in breast cancer Permalink https://escholarship.org/uc/item/1zq9j4nw Journal Molecular Cancer, 12(1) ISSN 1476-4598 Authors Ko, Jae-Hong Ko, Eun A Gu, Wanjun et al. Publication Date 2013-09-22 DOI http://dx.doi.org/10.1186/1476-4598-12-106 Peer reviewed eScholarship.org Powered by the California Digital Library University of California Ko et al. Molecular Cancer 2013, 12:106 http://www.molecular-cancer.com/content/12/1/106 RESEARCH Open Access Expression profiling of ion channel genes predicts clinical outcome in breast cancer Jae-Hong Ko1, Eun A Ko2, Wanjun Gu3, Inja Lim1, Hyoweon Bang1* and Tong Zhou4,5* Abstract Background: Ion channels play a critical role in a wide variety of biological processes, including the development of human cancer. However, the overall impact of ion channels on tumorigenicity in breast cancer remains controversial. Methods: We conduct microarray meta-analysis on 280 ion channel genes. We identify candidate ion channels that are implicated in breast cancer based on gene expression profiling. We test the relationship between the expression of ion channel genes and p53 mutation status, ER status, and histological tumor grade in the discovery cohort. A molecular signature consisting of ion channel genes (IC30) is identified by Spearman’s rank correlation test conducted between tumor grade and gene expression. A risk scoring system is developed based on IC30. We test the prognostic power of IC30 in the discovery and seven validation cohorts by both Cox proportional hazard regression and log-rank test.
    [Show full text]
  • Formation of COPI-Coated Vesicles at a Glance Eric C
    © 2018. Published by The Company of Biologists Ltd | Journal of Cell Science (2018) 131, jcs209890. doi:10.1242/jcs.209890 CELL SCIENCE AT A GLANCE Formation of COPI-coated vesicles at a glance Eric C. Arakel1 and Blanche Schwappach1,2,* ABSTRACT unresolved, this review attempts to refocus the perspectives of The coat protein complex I (COPI) allows the precise sorting of lipids the field. and proteins between Golgi cisternae and retrieval from the Golgi KEY WORDS: Arf1, ArfGAP, COPI, Coatomer, Golgi, Endoplasmic to the ER. This essential role maintains the identity of the early reticulum, Vesicle coat secretory pathway and impinges on key cellular processes, such as protein quality control. In this Cell Science at a Glance and accompanying poster, we illustrate the different stages of COPI- Introduction coated vesicle formation and revisit decades of research in the Vesicle coat proteins, such as the archetypal clathrin and the coat context of recent advances in the elucidation of COPI coat structure. protein complexes II and I (COPII and COPI, respectively) are By calling attention to an array of questions that have remained molecular machines with two central roles: enabling vesicle formation, and selecting protein and lipid cargo to be packaged within them. Thus, coat proteins fulfil a central role in the 1Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee homeostasis of the cell’s endomembrane system and are the basis 23, 37073 Göttingen, Germany. 2Max-Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany. of functionally segregated compartments. COPI operates in retrieval from the Golgi to the endoplasmic reticulum (ER) and in intra-Golgi *Author for correspondence ([email protected]) transport (Beck et al., 2009; Duden, 2003; Lee et al., 2004a; Spang, E.C.A., 0000-0001-7716-7149; B.S., 0000-0003-0225-6432 2009), and maintains ER- and Golgi-resident chaperones and enzymes where they belong.
    [Show full text]
  • Supplementary Figures 1-14 and Supplementary References
    SUPPORTING INFORMATION Spatial Cross-Talk Between Oxidative Stress and DNA Replication in Human Fibroblasts Marko Radulovic,1,2 Noor O Baqader,1 Kai Stoeber,3† and Jasminka Godovac-Zimmermann1* 1Division of Medicine, University College London, Center for Nephrology, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK. 2Insitute of Oncology and Radiology, Pasterova 14, 11000 Belgrade, Serbia 3Research Department of Pathology and UCL Cancer Institute, Rockefeller Building, University College London, University Street, London WC1E 6JJ, UK †Present Address: Shionogi Europe, 33 Kingsway, Holborn, London WC2B 6UF, UK TABLE OF CONTENTS 1. Supplementary Figures 1-14 and Supplementary References. Figure S-1. Network and joint spatial razor plot for 18 enzymes of glycolysis and the pentose phosphate shunt. Figure S-2. Correlation of SILAC ratios between OXS and OAC for proteins assigned to the SAME class. Figure S-3. Overlap matrix (r = 1) for groups of CORUM complexes containing 19 proteins of the 49-set. Figure S-4. Joint spatial razor plots for the Nop56p complex and FIB-associated complex involved in ribosome biogenesis. Figure S-5. Analysis of the response of emerin nuclear envelope complexes to OXS and OAC. Figure S-6. Joint spatial razor plots for the CCT protein folding complex, ATP synthase and V-Type ATPase. Figure S-7. Joint spatial razor plots showing changes in subcellular abundance and compartmental distribution for proteins annotated by GO to nucleocytoplasmic transport (GO:0006913). Figure S-8. Joint spatial razor plots showing changes in subcellular abundance and compartmental distribution for proteins annotated to endocytosis (GO:0006897). Figure S-9. Joint spatial razor plots for 401-set proteins annotated by GO to small GTPase mediated signal transduction (GO:0007264) and/or GTPase activity (GO:0003924).
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. 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. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. 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 The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • Kir4.1 May Represent a Novel Therapeutic Target for Diabetic Retinopathy (Review)
    EXPERIMENTAL AND THERAPEUTIC MEDICINE 22: 1021, 2021 Kir4.1 may represent a novel therapeutic target for diabetic retinopathy (Review) XIAOYU LI1,2, JIAJUN LV1,2, JIAZHI LI2 and XIANG REN1 1Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044; 2Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China Received April 6, 2021; Accepted May 28, 2021 DOI: 10.3892/etm.2021.10453 Abstract. As the major cause of irreversible loss of vision in individuals with diabetes worldwide, ~10% of which have adults, diabetic retinopathy (DR) is one of the most serious severe visual impairment and 2% of them are blind. It is complications of diabetes. The imbalance of the retinal micro‑ expected that the number of individuals at risk of vision loss environment and destruction of the blood‑retinal barrier have from DR will be double by 2030 (1). a significant role in the progression of DR. Inward rectifying Strategies to prevent or treat DR early have become a potassium channel 4.1 (Kir4.1) is located on Müller cells and research hotspot. An increasing number of studies have is closely related to potassium homeostasis, water balance and indicated that the occurrence of retinal neurodegenerative glutamate clearance in the whole retina. The present review changes in DR may be earlier than microvascular changes. discusses the functions of Kir4.1 in regulating the retinal Furthermore, both the proliferation of glial cells and the microenvironment and related biological mechanisms in DR. In damage of photoreceptor cells may occur at the beginning the future, Kir4.1 may represent a novel alternative therapeutic of the disease (2,3).
    [Show full text]
  • An Advance About the Genetic Causes of Epilepsy
    E3S Web of Conferences 271, 03068 (2021) https://doi.org/10.1051/e3sconf/202127103068 ICEPE 2021 An advance about the genetic causes of epilepsy Yu Sun1, a, *, †, Licheng Lu2, b, *, †, Lanxin Li3, c, *, †, Jingbo Wang4, d, *, † 1The School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801-3633, US 2High School Affiliated to Shanghai Jiao Tong University, Shanghai, 200441, China 3Applied Biology program, University of British Columbia, Vancouver, V6r3b1, Canada 4School of Chemical Machinery and Safety, Dalian University of Technology, Dalian, 116023, China †These authors contributed equally. Abstract: Human hereditary epilepsy has been found related to ion channel mutations in voltage-gated channels (Na+, K+, Ca2+, Cl-), ligand gated channels (GABA receptors), and G-protein coupled receptors, such as Mass1. In addition, some transmembrane proteins or receptor genes, including PRRT2 and nAChR, and glucose transporter genes, such as GLUT1 and SLC2A1, are also about the onset of epilepsy. The discovery of these genetic defects has contributed greatly to our understanding of the pathology of epilepsy. This review focuses on introducing and summarizing epilepsy-associated genes and related findings in recent decades, pointing out related mutant genes that need to be further studied in the future. 1 Introduction Epilepsy is a neurological disorder characterized by 2 Malfunction of Ion channel epileptic seizures caused by abnormal brain activity. 1 in Functional variation in voltage or ligand-gated ion 100 (50 million people) people are affected by symptoms channel mutations is a major cause of idiopathic epilepsy, of this disorder worldwide, with men, young children, and especially in rare genetic forms.
    [Show full text]
  • Rabbit Anti-Human KCNAB2 Polyclonal Antibody (DPABH-15471) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Rabbit Anti-Human KCNAB2 Polyclonal antibody (DPABH-15471) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Immunogen KCNAB2 fusion protein, sequence: MYPESTTGSPARLSLRQTGSPGMIYSTRYGSPKRQLQFYRNLGKSGLRVSCLGLGTWVTFGG QITDEMAEQLMTLAYDNGINLFDTAEVYAAGKAEVVLGNIIKKKGWRRSSLVITTKIFWGGKAETE RGLSRKHIIEGLKASLERLQLEYVDVVFANRPDPNTPMEETVRAMTHVINQGMAMYWGTSRWS SMEIMEAYSVARQFNLTPPICEQAEYHMFQREKVEVQLPELFHKIGVGAMTWSPLACGIVSGKY DSGIPPYSRASLKGYQWLKDKILSEEGRRQQAKLKELQAIAERLGCTLPQLAIAWCLRNEGVSSV LLGASNADQLMENIGAIQVRVRGPAGQRAHPSPSPVQCILPGSSCVPGSVLGTQDAPVNHQSC APGELAFQQEQT (1-395 aa encoded by BC110351) Isotype IgG Source/Host Rabbit Species Reactivity Human, Mouse, Rat Purification Antigen affinity purification Conjugate Unconjugated Applications WB, IHC, ELISA Positive Control mouse brain tissue, HepG2 cells Format Liquid Size 50 uL; 100 uL Buffer PBS with 0.02% sodium azide and 50% glycerol pH 7.3. Preservative 0.02% Sodium Azide Storage Store at -20°C. Aliquoting is unnecessary for -20°C storage. BACKGROUND Introduction Voltage-gated potassium (Kv) channels represent the most complex class of voltage-gated ion 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 channels from both functional and structural standpoints. Their diverse functions include regulating neurotransmitter release, heart rate, insulin secretion, neuronal excitability, epithelial electrolyte transport, smooth muscle contraction, and
    [Show full text]