Your browser doesn't support javascript.
loading
ASCC1 structures and bioinformatics reveal a novel helix-clasp-helix RNA-binding motif linked to a two-histidine phosphodiesterase.
Chinnam, Naga Babu; Thapar, Roopa; Arvai, Andrew S; Sarker, Altaf H; Soll, Jennifer M; Paul, Tanmoy; Syed, Aleem; Rosenberg, Daniel J; Hammel, Michal; Bacolla, Albino; Katsonis, Panagiotis; Asthana, Abhishek; Tsai, Miaw-Sheue; Ivanov, Ivaylo; Lichtarge, Olivier; Silverman, Robert H; Mosammaparast, Nima; Tsutakawa, Susan E; Tainer, John A.
Afiliación
  • Chinnam NB; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Thapar R; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Arvai AS; Integrative Structural & Computational Biology, The Scripps Research Institute, La Jolla, California, USA.
  • Sarker AH; Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
  • Soll JM; Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
  • Paul T; Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA.
  • Syed A; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Rosenberg DJ; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
  • Hammel M; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
  • Bacolla A; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Katsonis P; Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.
  • Asthana A; Department Cancer Biology, Cleveland Clinic Foundation, Lerner Research Institute, Cleveland, Ohio, USA.
  • Tsai MS; Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
  • Ivanov I; Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA.
  • Lichtarge O; Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.
  • Silverman RH; Department Cancer Biology, Cleveland Clinic Foundation, Lerner Research Institute, Cleveland, Ohio, USA.
  • Mosammaparast N; Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.
  • Tsutakawa SE; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA. Electronic address: setsutakawa@lbl.gov.
  • Tainer JA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA; Department of Cancer Biology, University of Texas MD Anderson Cance
J Biol Chem ; 300(6): 107368, 2024 Jun.
Article en En | MEDLINE | ID: mdl-38750793
ABSTRACT
Activating signal co-integrator complex 1 (ASCC1) acts with ASCC-ALKBH3 complex in alkylation damage responses. ASCC1 uniquely combines two evolutionarily ancient domains nucleotide-binding K-Homology (KH) (associated with regulating splicing, transcriptional, and translation) and two-histidine phosphodiesterase (PDE; associated with hydrolysis of cyclic nucleotide phosphate bonds). Germline mutations link loss of ASCC1 function to spinal muscular atrophy with congenital bone fractures 2 (SMABF2). Herein analysis of The Cancer Genome Atlas (TCGA) suggests ASCC1 RNA overexpression in certain tumors correlates with poor survival, Signatures 29 and 3 mutations, and genetic instability markers. We determined crystal structures of Alvinella pompejana (Ap) ASCC1 and Human (Hs) PDE domain revealing high-resolution details and features conserved over 500 million years of evolution. Extending our understanding of the KH domain Gly-X-X-Gly sequence motif, we define a novel structural Helix-Clasp-Helix (HCH) nucleotide binding motif and show ASCC1 sequence-specific binding to CGCG-containing RNA. The V-shaped PDE nucleotide binding channel has two His-Φ-Ser/Thr-Φ (HXT) motifs (Φ being hydrophobic) positioned to initiate cyclic phosphate bond hydrolysis. A conserved atypical active-site histidine torsion angle implies a novel PDE substrate. Flexible active site loop and arginine-rich domain linker appear regulatory. Small-angle X-ray scattering (SAXS) revealed aligned KH-PDE RNA binding sites with limited flexibility in solution. Quantitative evolutionary bioinformatic analyses of disease and cancer-associated mutations support implied functional roles for RNA binding, phosphodiesterase activity, and regulation. Collective results inform ASCC1's roles in transactivation and alkylation damage responses, its targeting by structure-based inhibitors, and how ASCC1 mutations may impact inherited disease and cancer.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Hidrolasas Diéster Fosfóricas Límite: Humans Idioma: En Revista: J Biol Chem Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Hidrolasas Diéster Fosfóricas Límite: Humans Idioma: En Revista: J Biol Chem Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos
...