Your browser doesn't support javascript.
loading
Proteome-wide analysis identifies plasma immune regulators of amyloid-beta progression.
Duggan, Michael R; Gomez, Gabriela T; Joynes, Cassandra M; Bilgel, Murat; Chen, Jingsha; Fattorelli, Nicola; Hohman, Timothy J; Mancuso, Renzo; Cordon, Jenifer; Castellano, Tonnar; Koran, Mary Ellen I; Candia, Julián; Lewis, Alexandria; Moghekar, Abhay; Ashton, Nicholas J; Kac, Przemyslaw R; Karikari, Thomas K; Blennow, Kaj; Zetterberg, Henrik; Martinez-Muriana, Anna; De Strooper, Bart; Thambisetty, Madhav; Ferrucci, Luigi; Gottesman, Rebecca F; Coresh, Josef; Resnick, Susan M; Walker, Keenan A.
Afiliação
  • Duggan MR; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
  • Gomez GT; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
  • Joynes CM; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
  • Bilgel M; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
  • Chen J; Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
  • Fattorelli N; VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium.
  • Hohman TJ; Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
  • Mancuso R; Microglia and Inflammation in Neurological Disorders Laboratory, Center for Molecular Neurology, Flanders Institute for Biotechnology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
  • Cordon J; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
  • Castellano T; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
  • Koran MEI; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
  • Candia J; Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
  • Lewis A; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
  • Moghekar A; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
  • Ashton NJ; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; NIHR Biomedical Research Center for Mental Health and Biomedical Research Unit for Dement
  • Kac PR; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden.
  • Karikari TK; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
  • Blennow K; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; ICM Institute, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, Fr
  • Zetterberg H; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, University College London Institute of Neur
  • Martinez-Muriana A; VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium.
  • De Strooper B; VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium; UK Dementia Research Institute, University College London, London, UK.
  • Thambisetty M; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
  • Ferrucci L; Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
  • Gottesman RF; Stroke Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
  • Coresh J; Departments of Population Health and Medicine, New York University Grossman School of Medicine, New York, NY, USA.
  • Resnick SM; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
  • Walker KA; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA. Electronic address: keenan.walker@nih.gov.
Brain Behav Immun ; 120: 604-619, 2024 Aug.
Article em En | MEDLINE | ID: mdl-38977137
ABSTRACT
While immune function is known to play a mechanistic role in Alzheimer's disease (AD), whether immune proteins in peripheral circulation influence the rate of amyloid-ß (Aß) progression - a central feature of AD - remains unknown. In the Baltimore Longitudinal Study of Aging, we quantified 942 immunological proteins in plasma and identified 32 (including CAT [catalase], CD36 [CD36 antigen], and KRT19 [keratin 19]) associated with rates of cortical Aß accumulation measured with positron emission tomography (PET). Longitudinal changes in a subset of candidate proteins also predicted Aß progression, and the mid- to late-life (20-year) trajectory of one protein, CAT, was associated with late-life Aß-positive status in the Atherosclerosis Risk in Communities (ARIC) study. Genetic variation that influenced plasma levels of CAT, CD36 and KRT19 predicted rates of Aß accumulation, including causal relationships with Aß PET levels identified with two-sample Mendelian randomization. In addition to associations with tau PET and plasma AD biomarker changes, as well as expression patterns in human microglia subtypes and neurovascular cells in AD brain tissue, we showed that 31 % of candidate proteins were related to mid-life (20-year) or late-life (8-year) dementia risk in ARIC. Our findings reveal plasma proteins associated with longitudinal Aß accumulation, and identify specific peripheral immune mediators that may contribute to the progression of AD pathophysiology.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Biomarcadores / Peptídeos beta-Amiloides / Progressão da Doença / Tomografia por Emissão de Pósitrons / Doença de Alzheimer Limite: Aged / Aged80 / Female / Humans / Male / Middle aged Idioma: En Revista: Brain Behav Immun Assunto da revista: ALERGIA E IMUNOLOGIA / CEREBRO / PSICOFISIOLOGIA Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Biomarcadores / Peptídeos beta-Amiloides / Progressão da Doença / Tomografia por Emissão de Pósitrons / Doença de Alzheimer Limite: Aged / Aged80 / Female / Humans / Male / Middle aged Idioma: En Revista: Brain Behav Immun Assunto da revista: ALERGIA E IMUNOLOGIA / CEREBRO / PSICOFISIOLOGIA Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos