Your browser doesn't support javascript.
loading
The human brainome: network analysis identifies HSPA2 as a novel Alzheimer’s disease target.
Petyuk, Vladislav A; Chang, Rui; Ramirez-Restrepo, Manuel; Beckmann, Noam D; Henrion, Marc Y R; Piehowski, Paul D; Zhu, Kuixi; Wang, Sven; Clarke, Jennifer; Huentelman, Matthew J; Xie, Fang; Andreev, Victor; Engel, Anzhelika; Guettoche, Toumy; Navarro, Loida; De Jager, Philip; Schneider, Julie A; Morris, Christopher M; McKeith, Ian G; Perry, Robert H; Lovestone, Simon; Woltjer, Randall L; Beach, Thomas G; Sue, Lucia I; Serrano, Geidy E; Lieberman, Andrew P; Albin, Roger L; Ferrer, Isidre; Mash, Deborah C; Hulette, Christine M; Ervin, John F; Reiman, Eric M; Hardy, John A; Bennett, David A; Schadt, Eric; Smith, Richard D; Myers, Amanda J.
Afiliação
  • Petyuk VA; Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA.
  • Chang R; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
  • Ramirez-Restrepo M; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.
  • Beckmann ND; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
  • Henrion MYR; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
  • Piehowski PD; Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA.
  • Zhu K; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
  • Wang S; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
  • Clarke J; Food Science and Technology Department, University of Nebraska-Lincoln, Lincoln, NE, USA.
  • Huentelman MJ; Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA.
  • Xie F; Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA.
  • Andreev V; Arbor Research Collaborative for Health, 340 E Huron St # 300, Ann Arbor, MI, USA.
  • Engel A; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.
  • Guettoche T; Roche Sequencing, 4300 Hacienda Drive, Pleasanton, CA, USA.
  • Navarro L; Roche Sequencing, 4300 Hacienda Drive, Pleasanton, CA, USA.
  • De Jager P; Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA.
  • Schneider JA; New York Genome Center, New York NY, USA.
  • Morris CM; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
  • McKeith IG; Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA.
  • Perry RH; Newcastle Brain Tissue Resource, Institute of Neuroscience, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK.
  • Lovestone S; NIHR Biomedical Research Centre, Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK.
  • Woltjer RL; Neuropathology and Cellular Pathology, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, UK.
  • Beach TG; University of Oxford, Medical Sciences Division, Department of Psychiatry, Warneford Hospital, Oxford, UK.
  • Sue LI; Neuropathology Core of the Layton Aging and Alzheimer’s Disease Center, Oregon Health and Science University, Portland, OR, USA.
  • Serrano GE; Banner Sun Health Research Institute, Sun City, AZ, USA.
  • Lieberman AP; Banner Sun Health Research Institute, Sun City, AZ, USA.
  • Albin RL; Banner Sun Health Research Institute, Sun City, AZ, USA.
  • Ferrer I; Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
  • Mash DC; Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
  • Hulette CM; Geriatrics Research, Education, and Clinical Center, VAAAHS, Ann Arbor, MI, USA.
  • Ervin JF; Department of Pathology and Experimental Therapeutics, University of Barcelona; CIBERNED; Hospitalet de Llobregat, Spain.
  • Reiman EM; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA.
  • Hardy JA; Department of Pathology, Division of Neuropathology, Duke University Medical Center, Durham, NC, USA.
  • Bennett DA; Kathleen Price Bryan Brain Bank, Department of Medicine, Division of Neurology, Duke University, Durham, NC, USA.
  • Schadt E; The Arizona Alzheimer’s Consortium, Phoenix, Arizona, USA.
  • Smith RD; Banner Alzheimer’s Institute, Phoenix, Arizona, USA.
  • Myers AJ; Department of Molecular Neuroscience and Reta Lila Research Laboratories, University College London Institute of Neurology, London, UK.
Brain ; 141(9): 2721-2739, 2018 09 01.
Article em En | MEDLINE | ID: mdl-30137212
ABSTRACT
Our hypothesis is that changes in gene and protein expression are crucial to the development of late-onset Alzheimer’s disease. Previously we examined how DNA alleles control downstream expression of RNA transcripts and how those relationships are changed in late-onset Alzheimer’s disease. We have now examined how proteins are incorporated into networks in two separate series and evaluated our outputs in two different cell lines. Our pipeline included the following

steps:

(i) predicting expression quantitative trait loci; (ii) determining differential expression; (iii) analysing networks of transcript and peptide relationships; and (iv) validating effects in two separate cell lines. We performed all our analysis in two separate brain series to validate effects. Our two series included 345 samples in the first set (177 controls, 168 cases; age range 65–105; 58% female; KRONOSII cohort) and 409 samples in the replicate set (153 controls, 141 cases, 115 mild cognitive impairment; age range 66–107; 63% female; RUSH cohort). Our top target is heat shock protein family A member 2 (HSPA2), which was identified as a key driver in our two datasets. HSPA2 was validated in two cell lines, with overexpression driving further elevation of amyloid-β40 and amyloid-β42 levels in APP mutant cells, as well as significant elevation of microtubule associated protein tau and phosphorylated-tau in a modified neuroglioma line. This work further demonstrates that studying changes in gene and protein expression is crucial to understanding late onset disease and further nominates HSPA2 as a specific key regulator of late-onset Alzheimer’s disease processes.10.1093/brain/awy215_video1awy215media15824729224001.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteínas de Choque Térmico HSP70 / Doença de Alzheimer Limite: Aged / Aged80 / Female / Humans / Male Idioma: En Revista: Brain Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteínas de Choque Térmico HSP70 / Doença de Alzheimer Limite: Aged / Aged80 / Female / Humans / Male Idioma: En Revista: Brain Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Estados Unidos