Analysis of Genomic Alterations in Malignant Melanoma”

Total Page:16

File Type:pdf, Size:1020Kb

Analysis of Genomic Alterations in Malignant Melanoma” Dissertation zur Erlangung des Doktorgrades der Naturwissenschaften der Fakultät für Biologie der Ludwig-Maximilians-Universität München Analysis of genomic alterations in malignant melanoma vorgelegt von Henrike Körner am 02.05.2007 Die Arbeit wurde am Max-Planck Institut für Biochemie (Martinsried) in der NWG Molekulare Onkologie angefertigt. First examiner: PD Dr. Heiko Hermeking Second examiner: Prof. Dr. Thomas Cremer Additional examiners: Prof. Dr. Michael Boshart Prof. Dr. Heinrich Leonhardt Date of the oral examination: 05.12.2007 Erklärung Hiermit erkläre ich, dass ich die vorliegende Dissertation selbstständig verfasst habe und keine anderen als die von mir angegebenen Quellen und Hilfsmittel benutzt habe. Sämtliche Experimente sind von mir selbst durchgeführt worden, außer wenn explizit auf dritte verwiesen wird. Ferner erkläre ich, dass ich nicht anderweitig versucht habe, eine Dissertation oder Teile einer Dissertation einzureichen bzw. einer Prüfungskommission vorzulegen oder mich einer Doktorprüfung zu unterziehen. München, den 07.12.07 Henrike Körner TABLE OF CONTENT TABLE OF CONTENT 1 INTRODUCTION..................................................................................................1 1.1 Cutaneous malignant melanoma...................................................................1 1.1.1 Germ line mutations in melanoma ..........................................................3 1.1.1.1 CDKN2A ..........................................................................................3 1.1.1.2 CDK4 ...............................................................................................5 1.1.2 Pigmentary traits.....................................................................................6 1.1.3 Somatic mutations in melanoma.............................................................7 1.1.3.1 Mitogen-activated protein kinase signaling pathway........................7 1.1.3.2 HGF/SF-MET signaling pathway .....................................................8 1.1.3.3 PTEN-AKT signaling pathway..........................................................8 1.2 Methods for cytogenetic and whole-genome analyses..................................9 1.2.1 Fluorescence in situ hybridization.........................................................10 1.2.2 Comparative genomic hybridization......................................................11 1.2.3 Digital karyotyping ................................................................................12 1.3 Dystrophin ...................................................................................................15 1.3.1 DMD gene and isoforms.......................................................................15 1.3.2 Dystrophin protein.................................................................................17 1.3.3 Duchenne and Becker Muscular Dystrophies.......................................18 1.3.4 Mutations in DMD .................................................................................19 1.3.5 Dystrophin-glycoprotein complex..........................................................20 2 AIM OF THE STUDY .........................................................................................22 3 MATERIALS ......................................................................................................23 3.1 Chemicals ...................................................................................................23 3.2 Enzymes .....................................................................................................24 3.3 Commercial kits and other materials ...........................................................24 3.4 Buffers and stock solutions .........................................................................25 3.5 Antibodies ...................................................................................................28 3.5.1 Primary antibodies................................................................................28 3.5.2 Secondary antibodies ...........................................................................29 3.6 DNA constructs ...........................................................................................29 3.7 Oligonucleotides..........................................................................................30 I TABLE OF CONTENT 3.8 Bacteria strains ...........................................................................................31 3.9 Cell lines and cell culture media..................................................................32 3.10 Equipment...................................................................................................32 4 METHODS .........................................................................................................33 4.1 Chromosome metaphase spreads ..............................................................33 4.2 Comparative genomic hybridization ............................................................33 4.3 7-Fluor M-FISH ...........................................................................................34 4.4 Digital karyotyping.......................................................................................35 4.5 Isolation of genomic DNA from cell lines and paraffin-embedded tissue sections.......................................................................................................40 4.6 RNA isolation and RT reaction ....................................................................40 4.7 RT-PCR analysis.........................................................................................40 4.8 qPCR analysis.............................................................................................41 4.9 PCR analysis of genomic DNA....................................................................41 4.10 DNA sequencing .........................................................................................42 4.11 Western blot analysis ..................................................................................42 4.12 Generation of stable cell lines .....................................................................42 4.13 Cell growth in matrigel.................................................................................43 4.14 Cell migration assays ..................................................................................44 4.15 Cell proliferation assay................................................................................44 4.16 Analysis of apoptosis by flow cytometry......................................................45 4.17 Senescence-associated β-galactosidase staining.......................................45 4.18 Immunofluorescence staining and confocal microscopy .............................46 4.19 Statistical analysis.......................................................................................46 5 RESULTS ..........................................................................................................48 5.1 Characterization of malignant melanoma cell lines .....................................48 5.1.1 General characteristics.........................................................................48 5.1.2 M-FISH analysis of malignant melanoma cell lines...............................49 5.1.3 CGH analysis of malignant melanoma cell lines...................................58 5.2 Digital karyotyping analysis of malignant melanoma cell lines ....................63 5.2.1 Validation of the digital karyotyping results...........................................63 5.3 Analysis of DMD in malignant melanoma....................................................67 II TABLE OF CONTENT 5.3.1 Deletion of DMD in malignant melanoma cell lines...............................67 5.3.2 Expression of DMD isoforms in primary melanocytes...........................68 5.3.3 Expression of dystrophin isoforms in melanoma cell lines....................70 5.3.4 Functional analysis of dystrophin..........................................................75 5.3.4.1 Analysis of the components of the dystrophin-glycoprotein complex .......................................................................................................75 5.3.4.2 Knockdown of DMD in melanoma cell lines...................................81 5.3.4.3 Re-expression of dystrophin in melanoma cell lines ......................83 6 DISCUSSION.....................................................................................................86 6.1 Cytogenetic characterization of melanoma cell lines...................................86 6.2 Digital karyotyping of melanoma cell lines...................................................89 6.3 Down-regulation of DMD in melanoma cell lines.........................................90 6.4 The dystrophin-glycoprotein complex in melanocytes and melanoma cell lines.............................................................................................................93 7 SUMMARY.........................................................................................................96 8 REFERENCES...................................................................................................98 9 ABBREVIATIONS ...........................................................................................130 10 APPENDIX.......................................................................................................132 11 PUBLICATION LIST........................................................................................137
Recommended publications
  • 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]
  • The Biomarkers of Key Mirnas and Target Genes Associated with Acute Myocardial Infarction
    The biomarkers of key miRNAs and target genes associated with acute myocardial infarction Qi Wang1, Bingyan Liu2,3, Yuanyong Wang4, Baochen Bai1, Tao Yu3 and Xian–ming Chu1,5 1 Department of Cardiology, The Affiliated hospital of Qingdao University, Qingdao, China 2 School of Basic Medicine, Qingdao University, Qingdao, China 3 Institute for Translational Medicine, Qingdao University, Qingdao, China 4 Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China 5 Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China ABSTRACT Background. Acute myocardial infarction (AMI) is considered one of the most prominent causes of death from cardiovascular disease worldwide. Knowledge of the molecular mechanisms underlying AMI remains limited. Accurate biomarkers are needed to predict the risk of AMI and would be beneficial for managing the incidence rate. The gold standard for the diagnosis of AMI, the cardiac troponin T (cTnT) assay, requires serial testing, and the timing of measurement with respect to symptoms affects the results. As attractive candidate diagnostic biomarkers in AMI, circulating microRNAs (miRNAs) are easily detectable, generally stable and tissue specific. Methods. The Gene Expression Omnibus (GEO) database was used to compare miRNA expression between AMI and control samples, and the interactions between miRNAs and mRNAs were analysed for expression and function. Furthermore, a protein-protein interaction (PPI) network was constructed. The miRNAs identified in the bioinformatic analysis were verified by RT-qPCR in an H9C2 cell line. The miRNAs in plasma samples from patients with AMI (n D 11) and healthy controls (n D 11) were used to construct Submitted 23 December 2019 receiver operating characteristic (ROC) curves to evaluate the clinical prognostic value Accepted 14 April 2020 of the identified miRNAs.
    [Show full text]
  • Individual Protomers of a G Protein-Coupled Receptor Dimer Integrate Distinct Functional Modules
    OPEN Citation: Cell Discovery (2015) 1, 15011; doi:10.1038/celldisc.2015.11 © 2015 SIBS, CAS All rights reserved 2056-5968/15 ARTICLE www.nature.com/celldisc Individual protomers of a G protein-coupled receptor dimer integrate distinct functional modules Nathan D Camp1, Kyung-Soon Lee2, Jennifer L Wacker-Mhyre2, Timothy S Kountz2, Ji-Min Park2, Dorathy-Ann Harris2, Marianne Estrada2, Aaron Stewart2, Alejandro Wolf-Yadlin1, Chris Hague2 1Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA; 2Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA Recent advances in proteomic technology reveal G-protein-coupled receptors (GPCRs) are organized as large, macromolecular protein complexes in cell membranes, adding a new layer of intricacy to GPCR signaling. We previously reported the α1D-adrenergic receptor (ADRA1D)—a key regulator of cardiovascular, urinary and CNS function—binds the syntrophin family of PDZ domain proteins (SNTA, SNTB1, and SNTB2) through a C-terminal PDZ ligand inter- action, ensuring receptor plasma membrane localization and G-protein coupling. To assess the uniqueness of this novel GPCR complex, 23 human GPCRs containing Type I PDZ ligands were subjected to TAP/MS proteomic analysis. Syntrophins did not interact with any other GPCRs. Unexpectedly, a second PDZ domain protein, scribble (SCRIB), was detected in ADRA1D complexes. Biochemical, proteomic, and dynamic mass redistribution analyses indicate syntrophins and SCRIB compete for the PDZ ligand, simultaneously exist within an ADRA1D multimer, and impart divergent pharmacological properties to the complex. Our results reveal an unprecedented modular dimeric architecture for the ADRA1D in the cell membrane, providing unexpected opportunities for fine-tuning receptor function through novel protein interactions in vivo, and for intervening in signal transduction with small molecules that can stabilize or disrupt unique GPCR:PDZ protein interfaces.
    [Show full text]
  • Supplementary Table 1
    Supplementary Table 1. Large-scale quantitative phosphoproteomic profiling was performed on paired vehicle- and hormone-treated mTAL-enriched suspensions (n=3). A total of 654 unique phosphopeptides corresponding to 374 unique phosphoproteins were identified. The peptide sequence, phosphorylation site(s), and the corresponding protein name, gene symbol, and RefSeq Accession number are reported for each phosphopeptide identified in any one of three experimental pairs. For those 414 phosphopeptides that could be quantified in all three experimental pairs, the mean Hormone:Vehicle abundance ratio and corresponding standard error are also reported. Peptide Sequence column: * = phosphorylated residue Site(s) column: ^ = ambiguously assigned phosphorylation site Log2(H/V) Mean and SE columns: H = hormone-treated, V = vehicle-treated, n/a = peptide not observable in all 3 experimental pairs Sig. column: * = significantly changed Log 2(H/V), p<0.05 Log (H/V) Log (H/V) # Gene Symbol Protein Name Refseq Accession Peptide Sequence Site(s) 2 2 Sig. Mean SE 1 Aak1 AP2-associated protein kinase 1 NP_001166921 VGSLT*PPSS*PK T622^, S626^ 0.24 0.95 PREDICTED: ATP-binding cassette, sub-family A 2 Abca12 (ABC1), member 12 XP_237242 GLVQVLS*FFSQVQQQR S251^ 1.24 2.13 3 Abcc10 multidrug resistance-associated protein 7 NP_001101671 LMT*ELLS*GIRVLK T464, S468 -2.68 2.48 4 Abcf1 ATP-binding cassette sub-family F member 1 NP_001103353 QLSVPAS*DEEDEVPVPVPR S109 n/a n/a 5 Ablim1 actin-binding LIM protein 1 NP_001037859 PGSSIPGS*PGHTIYAK S51 -3.55 1.81 6 Ablim1 actin-binding
    [Show full text]
  • Analysis of the Dystrophin Interactome
    Analysis of the dystrophin interactome Dissertation In fulfillment of the requirements for the degree “Doctor rerum naturalium (Dr. rer. nat.)” integrated in the International Graduate School for Myology MyoGrad in the Department for Biology, Chemistry and Pharmacy at the Freie Universität Berlin in Cotutelle Agreement with the Ecole Doctorale 515 “Complexité du Vivant” at the Université Pierre et Marie Curie Paris Submitted by Matthew Thorley born in Scunthorpe, United Kingdom Berlin, 2016 Supervisor: Simone Spuler Second examiner: Sigmar Stricker Date of defense: 7th December 2016 Dedicated to My mother, Joy Thorley My father, David Thorley My sister, Alexandra Thorley My fiancée, Vera Sakhno-Cortesi Acknowledgements First and foremost, I would like to thank my supervisors William Duddy and Stephanie Duguez who gave me this research opportunity. Through their combined knowledge of computational and practical expertise within the field and constant availability for any and all assistance I required, have made the research possible. Their overarching support, approachability and upbeat nature throughout, while granting me freedom have made this year project very enjoyable. The additional guidance and supported offered by Matthias Selbach and his team whenever required along with a constant welcoming invitation within their lab has been greatly appreciated. I thank MyoGrad for the collaboration established between UPMC and Freie University, creating the collaboration within this research project possible, and offering research experience in both the Institute of Myology in Paris and the Max Delbruck Centre in Berlin. Vital to this process have been Gisele Bonne, Heike Pascal, Lidia Dolle and Susanne Wissler who have aided in the often complex processes that I am still not sure I fully understand.
    [Show full text]
  • Supplementary Material
    Supplementary material: Figure S1. Protein-Protein Interactions of the 127 Dysregulated proteins in brain tissues following IONP exposure. The graph was generated from STRING database. 125 nodes, 183 edges, 2.93 Average node degree, 0.411 Avg. local clustering coefficient, 122 Expected number of edges, PPI enrichment p-value 1.83e-07. 1 Figure S2. Protein-Protein Interactions of the 66 Dysregulated proteins in liver tissues following IONP exposure. The graph was generated from STRING database. 61 nodes, 103 edges, 3.38 Average node degree, 0.412 Avg. local clustering coefficient, 22 Expected number of edges, PPI enrichment p-value < 1.0e-16. 2 Figure S3. Protein-Protein Interactions of the 84 Dysregulated proteins in lung tissues following IONP exposure. The graph was generated from STRING database. 82 nodes, 125 edges, 3.05 Average node degree, 0.422 Avg. local clustering coefficient, 64 Expected number of edges, PPI enrichment p-value 1.07e-11. 3 Table S1. List of all dysregulated proteins in brain tissues following IONP exposure. A) Down regulated proteins and B) Upregulated proteins. Statistically significant iTRAQ ratios (p-value ratio and p-value sample≤0.05) for the 127 proteins that are dysregulated. A) Sequence Peptide Spectral P-Value P-Value Log 10 Accession Name Description Gene Coverage Ratio Count Count Ratio Sample Ratio % P00762 TRY1_RAT Anionic trypsin-1 Prss1 1 8 8.13 0.526 0.001 0.004 -0.279 P08592 A4_RAT Amyloid beta A4 protein App 1 2 1.17 0.546 0.010 0.004 -0.263 Q62696 DLG1_RAT Disks large homolog 1 Dlg1 1 1 1.32
    [Show full text]
  • Peptidome Analysis of Umbilical Cord Mesenchymal Stem Cell (Huc-MSC
    Wang et al. Stem Cell Research & Therapy (2020) 11:414 https://doi.org/10.1186/s13287-020-01931-0 RESEARCH Open Access Peptidome analysis of umbilical cord mesenchymal stem cell (hUC-MSC) conditioned medium from preterm and term infants Yu Wang1,2†, Lin Zhang1†, Yun Wu3, Rongping Zhu1, Yan Wang2, Yan Cao2, Wei Long4, Chenbo Ji2, Huaiyan Wang1* and Lianghui You 2* Abstract Background: The therapeutic role of mesenchymal stem cells (MSCs) has been widely confirmed in several animal models of premature infant diseases. Micromolecule peptides have shown promise for the treatment of premature infant diseases. However, the potential role of peptides secreted from MSCs has not been studied. The purpose of this study is to help to broaden the knowledge of the hUC-MSC secretome at the peptide level through peptidomic profile analysis. Methods: We used tandem mass tag (TMT) labeling technology followed by tandem mass spectrometry to compare the peptidomic profile of preterm and term umbilical cord MSC (hUC-MSC) conditioned medium (CM). Gene Ontology (GO) enrichment analysis and ingenuity pathway analysis (IPA) were conducted to explore the differentially expressed peptides by predicting the functions of their precursor proteins. To evaluate the effect of candidate peptides on human lung epithelial cells stimulated by hydrogen peroxide (H2O2), quantitative real-time PCR (qRT-PCR), western blot analysis, and enzyme-linked immunosorbent assay (ELISA) were, respectively, adopted to detect inflammatory cytokines (TNF-α, IL-1β, and IL-6) expression levels at the mRNA and protein levels. Results: A total of 131 peptides derived from 106 precursor proteins were differentially expressed in the preterm hUC-MSC CM compared with the term group, comprising 37 upregulated peptides and 94 downregulated peptides.
    [Show full text]
  • Role of PDZ-Binding Motif from West Nile Virus NS5 Protein on Viral
    www.nature.com/scientificreports OPEN Role of PDZ‑binding motif from West Nile virus NS5 protein on viral replication Emilie Giraud1*, Chloé Otero del Val2, Célia Caillet‑Saguy2, Nada Zehrouni2, Cécile Khou5, Joël Caillet4, Yves Jacob3, Nathalie Pardigon5 & Nicolas Wolf2 West Nile virus (WNV) is a Flavivirus, which can cause febrile illness in humans that may progress to encephalitis. Like any other obligate intracellular pathogens, Flaviviruses hijack cellular protein functions as a strategy for sustaining their life cycle. Many cellular proteins display globular domain known as PDZ domain that interacts with PDZ‑Binding Motifs (PBM) identifed in many viral proteins. Thus, cellular PDZ‑containing proteins are common targets during viral infection. The non‑structural protein 5 (NS5) from WNV provides both RNA cap methyltransferase and RNA polymerase activities and is involved in viral replication but its interactions with host proteins remain poorly known. In this study, we demonstrate that the C‑terminal PBM of WNV NS5 recognizes several human PDZ‑ containing proteins using both in vitro and in cellulo high‑throughput methods. Furthermore, we constructed and assayed in cell culture WNV replicons where the PBM within NS5 was mutated. Our results demonstrate that the PBM of WNV NS5 is important in WNV replication. Moreover, we show that knockdown of the PDZ‑containing proteins TJP1, PARD3, ARHGAP21 or SHANK2 results in the decrease of WNV replication in cells. Altogether, our data reveal that interactions between the PBM of NS5 and PDZ‑containing proteins afect West Nile virus replication. Arboviruses include numerous human and animal pathogens that are important global health threats responsible for arboviroses.
    [Show full text]
  • Genome-Wide Insights on Gastrointestinal Nematode
    www.nature.com/scientificreports OPEN Genome‑wide insights on gastrointestinal nematode resistance in autochthonous Tunisian sheep A. M. Ahbara1,2, M. Rouatbi3,4, M. Gharbi3,4, M. Rekik1, A. Haile1, B. Rischkowsky1 & J. M. Mwacharo1,5* Gastrointestinal nematode (GIN) infections have negative impacts on animal health, welfare and production. Information from molecular studies can highlight the underlying genetic mechanisms that enhance host resistance to GIN. However, such information often lacks for traditionally managed indigenous livestock. Here, we analysed 600 K single nucleotide polymorphism genotypes of GIN infected and non‑infected traditionally managed autochthonous Tunisian sheep grazing communal natural pastures. Population structure analysis did not fnd genetic diferentiation that is consistent with infection status. However, by contrasting the infected versus non‑infected cohorts using ROH, LR‑GWAS, FST and XP‑EHH, we identifed 35 candidate regions that overlapped between at least two methods. Nineteen regions harboured QTLs for parasite resistance, immune capacity and disease susceptibility and, ten regions harboured QTLs for production (growth) and meat and carcass (fatness and anatomy) traits. The analysis also revealed candidate regions spanning genes enhancing innate immune defence (SLC22A4, SLC22A5, IL‑4, IL‑13), intestinal wound healing/repair (IL‑4, VIL1, CXCR1, CXCR2) and GIN expulsion (IL‑4, IL‑13). Our results suggest that traditionally managed indigenous sheep have evolved multiple strategies that evoke and enhance GIN resistance and developmental stability. They confrm the importance of obtaining information from indigenous sheep to investigate genomic regions of functional signifcance in understanding the architecture of GIN resistance. Small ruminants (sheep and goats) make immense socio-economic and cultural contributions across the globe.
    [Show full text]
  • Entrez ID 1 Symbol 1 Entrez ID 2 Symbol 2 Data Source (R
    Supporting Information Table 4. List of human protein-protein interactons. Entrez ID 1 Symbol 1 Entrez ID 2 Symbol 2 Data Source (R: Rual et al; S: Stelzl et al; L: Literature curation) 1 A1BG 10321 CRISP3 L 2 A2M 259 AMBP L 2 A2M 348 APOE L 2 A2M 351 APP L 2 A2M 354 KLK3 L 2 A2M 567 B2M L 2 A2M 1508 CTSB L 2 A2M 1990 ELA1 L 2 A2M 3309 HSPA5 L 2 A2M 3553 IL1B L 2 A2M 3586 IL10 L 2 A2M 3931 LCAT L 2 A2M 3952 LEP L 2 A2M 4035 LRP1 L 2 A2M 4803 NGFB L 2 A2M 5047 PAEP L 2 A2M 7045 TGFBI L 2 A2M 8728 ADAM19 L 2 A2M 9510 ADAMTS1 L 2 A2M 10944 SMAP S 2 A2M 55729 ATF7IP L 9 NAT1 8260 ARD1A L 12 SERPINA3 351 APP L 12 SERPINA3 354 KLK3 L 12 SERPINA3 1215 CMA1 L 12 SERPINA3 1504 CTRB1 L 12 SERPINA3 1506 CTRL L 12 SERPINA3 1511 CTSG L 12 SERPINA3 1990 ELA1 L 12 SERPINA3 1991 ELA2 L 12 SERPINA3 2064 ERBB2 L 12 SERPINA3 2153 F5 L 12 SERPINA3 3817 KLK2 L 12 SERPINA3 4035 LRP1 L 12 SERPINA3 4485 MST1 L 12 SERPINA3 5422 POLA L 12 SERPINA3 64215 DNAJC1 L 14 AAMP 51497 TH1L S 15 AANAT 7534 YWHAZ L 18 ABAT 7915 ALDH5A1 L 19 ABCA1 335 APOA1 L 19 ABCA1 6645 SNTB2 L 19 ABCA1 8772 FADD L 20 ABCA2 55755 CDK5RAP2 L 22 ABCB7 2235 FECH L 23 ABCF1 3692 ITGB4BP S 24 ABCA4 1258 CNGB1 L 25 ABL1 27 ABL2 L 25 ABL1 472 ATM L 25 ABL1 613 BCR L 25 ABL1 718 C3 L 25 ABL1 867 CBL L 25 ABL1 1501 CTNND2 L 25 ABL1 2048 EPHB2 L 25 ABL1 2547 XRCC6 L 25 ABL1 2876 GPX1 L 25 ABL1 2885 GRB2 L 25 ABL1 3055 HCK L 25 ABL1 3636 INPPL1 L 25 ABL1 3716 JAK1 L 25 ABL1 4193 MDM2 L 25 ABL1 4690 NCK1 L 25 ABL1 4914 NTRK1 L 25 ABL1 5062 PAK2 L 25 ABL1 5295 PIK3R1 L 25 ABL1 5335 PLCG1 L 25 ABL1 5591
    [Show full text]
  • Α-Dystrobrevin-1 Recruits Α-Catulin to the Α1d- Adrenergic Receptor/Dystrophin-Associated Protein Complex Signalosome
    α-Dystrobrevin-1 recruits α-catulin to the α1D- adrenergic receptor/dystrophin-associated protein complex signalosome John S. Lyssanda, Jennifer L. Whitingb, Kyung-Soon Leea, Ryan Kastla, Jennifer L. Wackera, Michael R. Bruchasa, Mayumi Miyatakea, Lorene K. Langebergb, Charles Chavkina, John D. Scottb, Richard G. Gardnera, Marvin E. Adamsc, and Chris Haguea,1 Departments of aPharmacology and cPhysiology and Biophysics, University of Washington, Seattle, WA 98195; and bDepartment of Pharmacology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195 Edited by Robert J. Lefkowitz, Duke University Medical Center/Howard Hughes Medical Institute, Durham, NC, and approved October 29, 2010 (received for review July 22, 2010) α1D-Adrenergic receptors (ARs) are key regulators of cardiovascu- pression increases in patients with benign prostatic hypertrophy lar system function that increase blood pressure and promote vas- (12). Through proteomic screening, we discovered that α1D-ARs cular remodeling. Unfortunately, little information exists about are scaffolded to the dystrophin-associated protein complex the signaling pathways used by this important G protein-coupled (DAPC) via the anchoring protein syntrophin (10). Coexpression α “ α receptor (GPCR). We recently discovered that 1D-ARs form a sig- with syntrophins increases 1D-AR plasma membrane expression, nalosome” with multiple members of the dystrophin-associated drug binding, and activation of Gαq/11 signaling after agonist protein complex (DAPC) to become functionally expressed at the activation. Moreover, syntrophin knockout mice lose α1D-AR– plasma membrane and bind ligands. However, the molecular stimulated increases in blood pressure, demonstrating the im- α α mechanism by which the DAPC imparts functionality to the 1D- portance of these essential GIPs for 1D-AR function in vivo (10).
    [Show full text]
  • UC San Diego Electronic Theses and Dissertations
    UC San Diego UC San Diego Electronic Theses and Dissertations Title Biomechanical implications of muscle cell-ECM communication Permalink https://escholarship.org/uc/item/8qf1428m Author Meyer, Gretchen Ann Publication Date 2011 Peer reviewed|Thesis/dissertation eScholarship.org Powered by the California Digital Library University of California UNIVERSITY OF CALIFORNIA, SAN DIEGO Biomechanical Implications of Muscle Cell-ECM Communication A dissertation submitted in partial satisfaction of the requirements for the degree Doctor of Philosophy in Bioengineering by Gretchen Ann Meyer Committee in charge: Professor Richard L. Lieber, Chair Professor Andrew D. McCulloch, Co-Chair Professor Anne Hoger Professor Geert Schmid-Schonbein Professor Samuel R. Ward 2011 Copyright Gretchen Ann Meyer, 2011 All rights reserved. The dissertation of Gretchen Ann Meyer is approved, and it is acceptable in quality and form for publication on microfilm and electronically: Co-Chair Chair University of California, San Diego 2011 iii DEDICATION This work is dedicated to my parents whose constant and unconditional support has always made me feel invincible. And to my colleagues and friends whose light has guided me through this mysterious world of science. iv EPIGRAPH Muscle is a metaphor for life; sometimes we could all use a good stretch. v TABLE OF CONTENTS Signature Page . iii Dedication . iv Epigraph . .v Table of Contents . vi List of Figures . .x List of Tables . xiii Acknowledgements . xiv Vita........................................ xvi Abstract of the Dissertation . xviii Chapter 1 Introduction . .1 1.1 Structure and function in skeletal muscle . .1 1.1.1 Extracellular structure . .2 1.1.2 Intracellular structure . .3 1.2 Desmin in muscle physiology .
    [Show full text]