Zebrafish Ryanodine Receptors Gene Protein Transcript Protein Identity Identity Genome Location Exons Γ Γ Name Name (Bp) (Aa) (1) (2)

Total Page:16

File Type:pdf, Size:1020Kb

Zebrafish Ryanodine Receptors Gene Protein Transcript Protein Identity Identity Genome Location Exons Γ Γ Name Name (Bp) (Aa) (1) (2) The role of ryanodine receptors in development Wu, Houdini Ho-Tin The copyright of this thesis rests with the author and no quotation from it or information derived from it may be published without the prior written consent of the author For additional information about this publication click this link. http://qmro.qmul.ac.uk/jspui/handle/123456789/1329 Information about this research object was correct at the time of download; we occasionally make corrections to records, please therefore check the published record when citing. For more information contact [email protected] THE ROLE OF RYANODINE RECEPTORS IN DEVELOPMENT by HOUDINI HO-TIN WU A thesis submitted to The University of London for the degree of DOCTOR OF PHILOSOPHY School of Biological and Chemical Sciences Queen Mary, University of London, Mile End Road, London E1 4NS United Kingdom April 2011 1 ABSTRACT Calcium ions (Ca2+) are fundamental to the regulation of many cellular processes; however, the coordination of these signals during embryogenesis is not well understood. Ryanodine receptors (RyR) are a family of important intracellular ion channels that are responsible for the release of Ca2+ and they regulate the cytosolic Ca2+ concentration. Humans have three differentially expressed ryr genes (ryr1, ryr2 and ryr3) and mutations can cause both skeletal and cardiac diseases. Although the primary function of RyR is to mediate excitation-contraction coupling in muscle, they may also regulate Ca2+ signalling during developmental processes. The project has addressed the role of RyR during embryonic development, using the zebrafish as an in vivo vertebrate model. Five zebrafish RyR genes (ryr1a, ryr1b, ryr2a, ryr2b and ryr3) were characterised and a comprehensive overview of their spatial and temporal expression in the embryo was determined. At 24 hours post- fertilisation (hpf), ryr1a, ryr1b and ryr3 are expressed in the skeletal muscle, ryr2a in specific neuronal populations and ryr2b in the cardiac muscle. Semi-quantitative PCR data and wholemount in situ hybridisation revealed strong maternal expression of ryr3 during the cleavage and blastula periods and into adulthood. The early expression of the ryr3 gene suggests that this receptor functions during the initial stages of development; a role that has not been described previously. The functional significance of RyR3 during early embryogenesis was investigated in a loss-of- 2 function model using antisense morpholino oligonucleotides. The ryr3 specific knockdown experiments appeared to affect the establishment of embryonic axis prior to the segmentation periods (before 10 hpf). In addition, by 19 to 20 hpf ryr3 morphants failed to exhibit spontaneous muscle contractions and displayed a defect in neuromuscular development. In conclusion, this study has characterised the ryr genes and provided an overview on their temporal and spatial expression. The work provides evidence that ryr3 expression provides the Ca2+ vital for myofibrils organisation and that is required for the spontaneous movements during zebrafish embryonic development. The knowledge of RyR tissue distribution in zebrafish has provided a strong foundation for loss-of-function studies aimed at addressing their role in development. In the long term, the work will also facilitate more focused studies on disease. 3 DECLARATION This work recorded in this thesis was carried out in the School of Biological and Chemical Sciences at the Queen Mary College, University of London, United Kingdom, during the period of October 2007 to September 2010. The work is original except where acknowledged by reference. No portion of the work is being or has been submitted for any other qualification at any other universities. 4 ♥ For my parents I.I. & Margaret and my wife Oona ♥ The thesis is also dedicated to my family ♥ 5 ACKNOWLEDGEMENTS First and foremost I would like to thank my supervisor, Dr. Rachel Ashworth for giving me this opportunity to be her first Ph.D. student to gain gratifying fruitful experience in her laboratory. I am very grateful to her for allowing me to present my work at conferences on many occasions. I would like to extend my thanks to Rachel and my second supervisor, Dr. Caroline Brennan for all the patience, support and invaluable advices they have given me throughout my Ph.D. Their inspiration in science has not only given me the motivations during the project, but also set me an excellent example to pursue a life-long career in scientific research, which has taken me to this point. A BIG THANK YOU TO BOTH OF YOU! Importantly, I would like to thank my parents I.I. and Margaret, my grandmother Lam Wai-Ying, my ‘great grandparents’, my brother Andy and my wife Oona for all their endless love, support and encouragements, enabling me to achieve my ambitions and to maintain my sanity. I would also like to thank my parents-in-law Leo and Katherine, my brothers-in-law Eddy and Gee, my sisters-in-law Jane and Gigi, uncle Allain and auntie Pauline for their caring and support (and their kindness :P). Thank you to both Uncle Two and Auntie Two, who have always treated me like their family members and bought me to golfing (a sport that I could never imagine I would be playing) and this really has broaden my horizons. All you guys mean so much to me and I really cannot imagine how I could do this without you! Particularly, I would like to thank my beloved one, Oona, who always loves me and my family. My life has been fantastic since your presence (28th July, 2006 just to remind myself in case I forgot ) and I know it always is my biggest fortune to have you in my life. Thank you so much for always being so supportive and kind to me. So, please keep it up :P and thank you! 6 Furthermore, I would also like to express my sincere gratitude to my college, SBCS for the pre-doctoral research studentship that funds my tuition fees, living and research costs, as well as my travel expenses, which has given me the opportunity to attend scientific workshop. In addition, I am very grateful to the Central Research Fund and Physiological Society travel grants for financially supporting my project to generate isoform specific RyR antibodies and enabling me to travel around the globe to present my work at conferences. A special thanks to Maggie & Haidee for the administrative help, Emma for the computing support, Ray for printing me poster up to the deadlines :P and my panel members (Prof Ralf Stanewsky, Drs Robin Maytum & Lesley Dobson) for their constructive comments for my project. Additionally, I would like to thank all my friends in Birmingham, Cambridge, Milton Keynes, London and Hong Kong. Thank you, Drs Abayomi Ogunbayo & Lynn Dover (for giving me an early interest into molecular biology), Carly Nichols & Heather Callaway (for the fish and eggs all these years), Chun Ming Chan (for your long-term support and encouragements), Prof David Minnikin (for keeping Oona in Birmingham), Debbie Goode & Dr Paul Piccinelli (for the advices on bioinformatics), Derek, Leo & Timothy (for our precious friendship), Eagle (for bringing me and Oona together), Dr Frank Michelangeli (for providing me an early ambition to do research and your invaluable advices), Dr Heidi Welch (for the great subcloning project to practise my molecular biology techniques and your encouragement), Drs John Puddefoot & Stewart Barker (for lending me your Western blotting equipment), Johnny Lau & Robert Li (for sharing golfing and magic tips), Drs Manuela Lahne & Yaniv Hintis (for sharing your scientific advices and brilliant ideas), Dr Michela Egertova (for teaching me how to do cryostat sectioning), Dr Raju Tatituri (for giving me an early ambition to pursue a Ph.D.), Miral Parmar (for your big support and sharing many funny jokes and video clips with me), Rosina Tsang (for the best pastries in Milton Keynes), Drs Sarah Batt, Usha Veeraraghavan & Natacha Veerapen (for your caring, support and taking good care of Oona for me while I’m in London). I really feel that it is my pleasure to have you all here throughout this journey. Thank you people!! ;) Finally, I have to say it always has been my pleasure to work on this fascinating project in Rachel’s lab and I hope my work will contribute to the community. 7 LIST OF ABBREVIATIONS ANOVA Analysis of variance AP Alkaline phosphatase ATP Adenosine 5’-triphosphate BCIP 5-bromo-4-chloro-3-indolyl phosphate BLAST Basic local alignment and search tool bp Base pairs Ca2+ Calcium ion [Ca2+] Concentration of calcium ions 2+ 2+ [Ca ]i Concentration of intracellular calcium ions (cytosolic Ca ) cAMP 3’, 5’-cyclic adenosine monophosphate cDNA Complementary DNA CICR Ca2+ induced Ca2+ release CNS Central nervous system Da Dalton DAB Diaminobenzidine DEPC Diethylpyrocarbonate DHPR Dihydropyridine receptor DICR Depolarisation induced Ca2+ release DIG Digoxygenin DMSO Dimethylsulfoxide DNA Deoxyribonucleic acid dNTPs Deoxynucleotidyl triphosphates DTT Dithiothreitol ECC Excitation-contraction coupling ECL Enhanced chemiluminescence EDTA Ethylenediaminetetraacetic acid EGTA Ethyleneglycoltetraacetic acid ELISA Enzyme linked immunosorbant assay 8 ER Endoplasmic reticulum EST Expressed cDNA sequence tag gDNA Genomic DNA HEPES N-[2-hydroxyethyl] piperazine-N’-[2-ethane sulphonic acid] hpf Hours post fertilisation HRP Horseradish peroxidase HS Horizontal myoseptum ICC Immunocytochemistry IgG Immunoglobulin G IHC Immunohistochemistry IP3 Inositol-1,4,5-trisphosphate IP3R IP3 receptor kb Kilo base pairs (103) kDa Kilo Dalton (103) MO Morpholino oligonucleotides
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]
  • Table 2. Significant
    Table 2. Significant (Q < 0.05 and |d | > 0.5) transcripts from the meta-analysis Gene Chr Mb Gene Name Affy ProbeSet cDNA_IDs d HAP/LAP d HAP/LAP d d IS Average d Ztest P values Q-value Symbol ID (study #5) 1 2 STS B2m 2 122 beta-2 microglobulin 1452428_a_at AI848245 1.75334941 4 3.2 4 3.2316485 1.07398E-09 5.69E-08 Man2b1 8 84.4 mannosidase 2, alpha B1 1416340_a_at H4049B01 3.75722111 3.87309653 2.1 1.6 2.84852656 5.32443E-07 1.58E-05 1110032A03Rik 9 50.9 RIKEN cDNA 1110032A03 gene 1417211_a_at H4035E05 4 1.66015788 4 1.7 2.82772795 2.94266E-05 0.000527 NA 9 48.5 --- 1456111_at 3.43701477 1.85785922 4 2 2.8237185 9.97969E-08 3.48E-06 Scn4b 9 45.3 Sodium channel, type IV, beta 1434008_at AI844796 3.79536664 1.63774235 3.3 2.3 2.75319499 1.48057E-08 6.21E-07 polypeptide Gadd45gip1 8 84.1 RIKEN cDNA 2310040G17 gene 1417619_at 4 3.38875643 1.4 2 2.69163229 8.84279E-06 0.0001904 BC056474 15 12.1 Mus musculus cDNA clone 1424117_at H3030A06 3.95752801 2.42838452 1.9 2.2 2.62132809 1.3344E-08 5.66E-07 MGC:67360 IMAGE:6823629, complete cds NA 4 153 guanine nucleotide binding protein, 1454696_at -3.46081884 -4 -1.3 -1.6 -2.6026947 8.58458E-05 0.0012617 beta 1 Gnb1 4 153 guanine nucleotide binding protein, 1417432_a_at H3094D02 -3.13334396 -4 -1.6 -1.7 -2.5946297 1.04542E-05 0.0002202 beta 1 Gadd45gip1 8 84.1 RAD23a homolog (S.
    [Show full text]
  • Transcriptomic Analysis of Native Versus Cultured Human and Mouse Dorsal Root Ganglia Focused on Pharmacological Targets Short
    bioRxiv preprint doi: https://doi.org/10.1101/766865; this version posted September 12, 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-ND 4.0 International license. Transcriptomic analysis of native versus cultured human and mouse dorsal root ganglia focused on pharmacological targets Short title: Comparative transcriptomics of acutely dissected versus cultured DRGs Andi Wangzhou1, Lisa A. McIlvried2, Candler Paige1, Paulino Barragan-Iglesias1, Carolyn A. Guzman1, Gregory Dussor1, Pradipta R. Ray1,#, Robert W. Gereau IV2, # and Theodore J. Price1, # 1The University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson, TX, 75080, USA 2Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine # corresponding authors [email protected], [email protected] and [email protected] Funding: NIH grants T32DA007261 (LM); NS065926 and NS102161 (TJP); NS106953 and NS042595 (RWG). The authors declare no conflicts of interest Author Contributions Conceived of the Project: PRR, RWG IV and TJP Performed Experiments: AW, LAM, CP, PB-I Supervised Experiments: GD, RWG IV, TJP Analyzed Data: AW, LAM, CP, CAG, PRR Supervised Bioinformatics Analysis: PRR Drew Figures: AW, PRR Wrote and Edited Manuscript: AW, LAM, CP, GD, PRR, RWG IV, TJP All authors approved the final version of the manuscript. 1 bioRxiv preprint doi: https://doi.org/10.1101/766865; this version posted September 12, 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.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Figure S1. Representative Report Generated by the Ion Torrent System Server for Each of the KCC71 Panel Analysis and Pcafusion Analysis
    Figure S1. Representative report generated by the Ion Torrent system server for each of the KCC71 panel analysis and PCaFusion analysis. (A) Details of the run summary report followed by the alignment summary report for the KCC71 panel analysis sequencing. (B) Details of the run summary report for the PCaFusion panel analysis. A Figure S1. Continued. Representative report generated by the Ion Torrent system server for each of the KCC71 panel analysis and PCaFusion analysis. (A) Details of the run summary report followed by the alignment summary report for the KCC71 panel analysis sequencing. (B) Details of the run summary report for the PCaFusion panel analysis. B Figure S2. Comparative analysis of the variant frequency found by the KCC71 panel and calculated from publicly available cBioPortal datasets. For each of the 71 genes in the KCC71 panel, the frequency of variants was calculated as the variant number found in the examined cases. Datasets marked with different colors and sample numbers of prostate cancer are presented in the upper right. *Significantly high in the present study. Figure S3. Seven subnetworks extracted from each of seven public prostate cancer gene networks in TCNG (Table SVI). Blue dots represent genes that include initial seed genes (parent nodes), and parent‑child and child‑grandchild genes in the network. Graphical representation of node‑to‑node associations and subnetwork structures that differed among and were unique to each of the seven subnetworks. TCNG, The Cancer Network Galaxy. Figure S4. REVIGO tree map showing the predicted biological processes of prostate cancer in the Japanese. Each rectangle represents a biological function in terms of a Gene Ontology (GO) term, with the size adjusted to represent the P‑value of the GO term in the underlying GO term database.
    [Show full text]
  • Transcriptional Control of Tissue-Resident Memory T Cell Generation
    Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2019 © 2019 Filip Cvetkovski All rights reserved ABSTRACT Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Tissue-resident memory T cells (TRM) are a non-circulating subset of memory that are maintained at sites of pathogen entry and mediate optimal protection against reinfection. Lung TRM can be generated in response to respiratory infection or vaccination, however, the molecular pathways involved in CD4+TRM establishment have not been defined. Here, we performed transcriptional profiling of influenza-specific lung CD4+TRM following influenza infection to identify pathways implicated in CD4+TRM generation and homeostasis. Lung CD4+TRM displayed a unique transcriptional profile distinct from spleen memory, including up-regulation of a gene network induced by the transcription factor IRF4, a known regulator of effector T cell differentiation. In addition, the gene expression profile of lung CD4+TRM was enriched in gene sets previously described in tissue-resident regulatory T cells. Up-regulation of immunomodulatory molecules such as CTLA-4, PD-1, and ICOS, suggested a potential regulatory role for CD4+TRM in tissues. Using loss-of-function genetic experiments in mice, we demonstrate that IRF4 is required for the generation of lung-localized pathogen-specific effector CD4+T cells during acute influenza infection. Influenza-specific IRF4−/− T cells failed to fully express CD44, and maintained high levels of CD62L compared to wild type, suggesting a defect in complete differentiation into lung-tropic effector T cells.
    [Show full text]
  • Yan Et Al. Supplementary Material
    SUPPLEMENTARY MATERIAL FOR: CELL ATLAS OF THE HUMAN FOVEA AND PERIPHERAL RETINA Wenjun Yan*, Yi-Rong Peng*, Tavé van Zyl*, Aviv Regev, Karthik Shekhar, Dejan Juric, and Joshua R, Sanes^ *Co-First authors ^Author for correspondence, [email protected] Figure S1 tSNE visualization showing contributions to cell types by batch for photoreceptors (a), horizontal cells (b), bipolar cells (c), amacrine cells (d), retinal ganglion cells (e) and non-neuronal cells (f). Each dot represents one cell. Colors distinguish retina samples. Source of each sample is shown in Table S1. Overall, batch eFFects were minimal. Figure S2 Violin and superimposed box plots showing expression of OPN4 in RGC clusters Figure S3 Heat maps showing expression patterns of disease genes by cell classes in the Fovea and periphery. Only genes expressed by more than 20% of cells in any individual class in either Fovea or peripheral cells are plotted. Table S1 Information on donors from whom retinal cells were obtained for scRNA-seq proFiling. Table S2 Publications reporting single cell or single nucleus profiling on cells from human retina. 1 Figure S1 a b c PR HC BP H1 H2F1 H2F2 H3 tSNE1 tSNE1 H4 tSNE1 H5 H9 H11 tSNE2 tSNE2 tSNE2 e AC f RGC g Non-neuronal tSNE1 tSNE1 tSNE1 tSNE2 tSNE2 tSNE2 Figure S2 OPN4 4 2 log Expression 0 MG-ON MG-OFF PG-OFF PG-ON hRGC5 hRGC6 hRGC7 hRGC8 hRGC9 hRGC10 hRGC11 hRGC12 Figure S3 Fovea Fovea Peripheral Peripheral Rods Cones BP HC AC RGC Muller Astro MicG Endo Rods Cones BP HC AC RGC Muller Astro MicG Endo ARL13B MAPK8IP3 EXOC6 LSM4 Expression
    [Show full text]
  • Stac Adaptor Proteins Regulate Trafficking and Function of Muscle
    Stac adaptor proteins regulate trafficking and function + of muscle and neuronal L-type Ca2 channels Alexander Polster, Stefano Perni, Hicham Bichraoui, and Kurt G. Beam1 Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Contributed by Kurt G. Beam, December 5, 2014 (sent for review November 11, 2014; reviewed by Bruce P. Bean and Terry P. Snutch) + Excitation–contraction (EC) coupling in skeletal muscle depends precisely regulated. As for other high-voltage activated Ca2 upon trafficking of CaV1.1, the principal subunit of the dihydropyr- channels, the auxiliary β-subunit is important for membrane traf- + idine receptor (DHPR) (L-type Ca2 channel), to plasma membrane ficking (5, 6). However, it is clear that an additional factor(s) is regions at which the DHPRs interact with type 1 ryanodine recep- also crucial. Specifically, CaV1.1 expresses poorly (7) or not at all tors (RyR1) in the sarcoplasmic reticulum. A distinctive feature of in mammalian cells that are not of muscle origin (e.g., tsA201 2+ this trafficking is that CaV1.1 expresses poorly or not at all in cells), whereas the closely related cardiac/neuronal L-type Ca mammalian cells that are not of muscle origin (e.g., tsA201 cells), channel, CaV1.2, is readily expressed in such cells (8–10). The in which all of the other nine CaV isoforms have been successfully absence of RyR1 seems insufficient to account for the difficulty expressed. Here, we tested whether plasma membrane trafficking of expressing Ca 1.1 in tsA201 cells because Ca 1.1 expresses of Ca 1.1 in tsA201 cells is promoted by the adapter protein Stac3, V V V well in dyspedic myotubes, albeit producing less current per because recent work has shown that genetic deletion of Stac3 in channel (3).
    [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]
  • Identification of Gene Expression and DNA Methylation of SERPINA5 and TIMP1 As Novel Prognostic Markers in Lower-Grade Gliomas
    Identification of gene expression and DNA methylation of SERPINA5 and TIMP1 as novel prognostic markers in lower-grade gliomas Wen-Jing Zeng1,2,3,4, Yong-Long Yang5, Zhi-Peng Wen1,2, Peng Chen1,2, Xiao-Ping Chen1,2 and Zhi-Cheng Gong3,4 1 Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China 2 Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, China 3 Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China 4 National Clinical Research Center for Geriatric Disorders (XIANGYA), Xiangya Hospital, Central South University, Changsha, Hunan, China 5 Department of Clinical Pharmacology Research Center, Changsha Carnation Geriatrics Hospital, Changsha, Hunan, China ABSTRACT Background. Lower-grade gliomas (LGGs) is characteristic with great difference in prognosis. Due to limited prognostic biomarkers, it is urgent to identify more molecular markers to provide a more objective and accurate tumor classification system for LGGs. Methods. In the current study, we performed an integrated analysis of gene expression data and genome-wide methylation data to determine novel prognostic genes and methylation sites in LGGs. Results. To determine genes that differentially expressed between 44 short-term survivors (<2 years) and 48 long-term survivors (≥2 years), we searched LGGs TCGA RNA-seq dataset and identified 106 differentially expressed genes. SERPINA5 and Submitted 11 October 2019 Accepted 9 May 2020 TIMP1 were selected for further study. Kaplan–Meier plots showed that SERPINA5 Published 3 June 2020 and TIMP1 expression were significantly correlated with overall survival (OS) and Corresponding authors relapse-free survival (RFS) in TCGA LGGs patients.
    [Show full text]
  • RYR3 Gene Polymorphisms and Cardiovascular Disease Outcomes in the Context of Antihypertensive Treatment
    The Pharmacogenomics Journal (2013) 13, 330 -- 334 & 2013 Macmillan Publishers Limited All rights reserved 1470-269X/13 www.nature.com/tpj ORIGINAL ARTICLE RYR3 gene polymorphisms and cardiovascular disease outcomes in the context of antihypertensive treatment AI Lynch1, MR Irvin1, E Boerwinkle2, BR Davis3, LK Vaughan4, CE Ford3, B Aissani1, JH Eckfeldt5, DK Arnett1 and S Shrestha1 Nearly one-third of adults in the United States have hypertension, which is associated with increased cardiovascular disease (CVD) morbidity and mortality. The goal of antihypertensive pharmacogenetic research is to enhance understanding of drug response based on the interaction of individual genetic architecture and antihypertensive therapy to improve blood pressure control and ultimately prevent CVD outcomes. In the context of the Genetics of Hypertension Associated Treatment study and using a case-only design, we examined whether single-nucleotide polymorphisms in RYR3 interact with four classes of antihypertensive drugs, particularly the calcium channel blocker amlodipine versus other classes, to modify the risk of coronary heart disease (CHD; fatal CHD and non-fatal myocardial infarction combined) and heart failure (HF) in high-risk hypertensive individuals. RYR3 mediates the mobilization of stored Ca þ 2 in cardiac and skeletal muscle to initiate muscle contraction. There was suggestive evidence of pharmacogenetic effects on HF, the strongest of which was for rs877087, with the smallest P-value ¼ 0.0005 for the codominant model when comparing amlodipine versus all other treatments. There were no pharmacogenetic effects observed for CHD. The findings reported here for the case-only analysis of the antihypertensive pharmacogenetic effect of RYR3 among 3058 CHD cases and 1940 HF cases show that a hypertensive patient’s genetic profile may help predict which medication(s) might better lower CVD risk.
    [Show full text]
  • Ion Channels 3 1
    r r r Cell Signalling Biology Michael J. Berridge Module 3 Ion Channels 3 1 Module 3 Ion Channels Synopsis Ion channels have two main signalling functions: either they can generate second messengers or they can function as effectors by responding to such messengers. Their role in signal generation is mainly centred on the Ca2 + signalling pathway, which has a large number of Ca2+ entry channels and internal Ca2+ release channels, both of which contribute to the generation of Ca2 + signals. Ion channels are also important effectors in that they mediate the action of different intracellular signalling pathways. There are a large number of K+ channels and many of these function in different + aspects of cell signalling. The voltage-dependent K (KV) channels regulate membrane potential and + excitability. The inward rectifier K (Kir) channel family has a number of important groups of channels + + such as the G protein-gated inward rectifier K (GIRK) channels and the ATP-sensitive K (KATP) + + channels. The two-pore domain K (K2P) channels are responsible for the large background K current. Some of the actions of Ca2 + are carried out by Ca2+-sensitive K+ channels and Ca2+-sensitive Cl − channels. The latter are members of a large group of chloride channels and transporters with multiple functions. There is a large family of ATP-binding cassette (ABC) transporters some of which have a signalling role in that they extrude signalling components from the cell. One of the ABC transporters is the cystic − − fibrosis transmembrane conductance regulator (CFTR) that conducts anions (Cl and HCO3 )and contributes to the osmotic gradient for the parallel flow of water in various transporting epithelia.
    [Show full text]