Plasma Glial Fibrillary Acidic Protein Levels Differ Along the Spectra of Amyloid Burden and Clinical Disease Stage.

BACKGROUND Measuring plasma glial fibrillary acidic protein (GFAP) alongside cortical amyloid-β (Aβ) may shed light on astrocytic changes in aging and Alzheimer's disease (AD). OBJECTIVE To examine associations between plasma GFAP and cortical Aβ deposition in older adults across the typical aging-to-AD dementia spectrum. METHODS We studied two independent samples from UCSF (Cohort 1, N = 50; Cohort 2, N = 37) covering the spectra of clinical severity (CDR Sum of Boxes; CDR-SB) and Aβ-PET burden. Aβ-PET was completed with either florbetapir or Pittsburgh Compound B and standardized uptake value ratios were converted to the Centiloid (CL) scale for analyses. All participants with CDR-SB >  0 were Aβ-PET positive, while clinically normal participants (CDR-SB = 0) were a mix of Aβ-PET positive and negative. Regression analyses evaluated main effect and interaction associations between plasma GFAP, Aβ-PET, and clinical severity. RESULTS In both cohorts, plasma GFAP increased linearly with Aβ-PET CLs in clinically normal older adults. In Cohort 2, which included participants with more severe clinical dysfunction and Aβ-PET burden, the association between Aβ and GFAP became curvilinear (inverted U-shape; quadratic model R2 change = 0.165, p = 0.009), and Aβ-PET interacted with CDR-SB (R2 change = 0.164, p = 0.007): older adults with intermediate functional impairment (CDR-SB = 0.5-4.0) showed a weak (negative) association between Aβ-PET CLs and plasma GFAP, while older adults with dementia (CDR-SB >  4.0) showed a strong, negative association of higher Aβ-PET CLs with lower plasma GFAP. CONCLUSION The relationship between astrocytic integrity and cortical Aβ may be highly dynamic, with linear, positive associations early in disease that diverge in more severe disease stages.

[1]  G. Kovacs Astroglia and Tau: New Perspectives , 2020, Frontiers in Aging Neuroscience.

[2]  Albert Hofman,et al.  Plasma tau, neurofilament light chain and amyloid-β levels and risk of dementia; a population-based cohort study , 2020, Brain : a journal of neurology.

[3]  G. Frisoni,et al.  Plasma glial fibrillary acidic protein is raised in progranulin-associated frontotemporal dementia , 2020, Journal of Neurology, Neurosurgery, and Psychiatry.

[4]  D. Harvey,et al.  Plasma biomarkers of astrocytic and neuronal dysfunction in early- and late-onset Alzheimer's disease , 2019, Alzheimer's & Dementia.

[5]  Nazanin Mirzaei,et al.  Ablation of reactive astrocytes exacerbates disease pathology in a model of Alzheimer's disease , 2019, Glia.

[6]  L. Tan,et al.  Neurofilament Light Chain in Cerebrospinal Fluid and Blood as a Biomarker for Neurodegenerative Diseases: A Systematic Review and Meta-Analysis. , 2019, Journal of Alzheimer's disease : JAD.

[7]  Philip S. Insel,et al.  Cerebrospinal fluid and plasma biomarker trajectories with increasing amyloid deposition in Alzheimer's disease , 2019, EMBO molecular medicine.

[8]  P. Calabresi,et al.  Neurofilament light chain as a biomarker in neurological disorders , 2019, Journal of Neurology, Neurosurgery, and Psychiatry.

[9]  Stephen F. Carter,et al.  Astrocyte Biomarkers in Alzheimer's Disease. , 2019, Trends in molecular medicine.

[10]  J. Kramer,et al.  A comparison of biofluid cytokine markers across platform technologies: Correspondence or divergence? , 2018, Cytokine.

[11]  J. Kassubek,et al.  Serum GFAP as a biomarker for disease severity in multiple sclerosis , 2018, Scientific Reports.

[12]  Prashanthi Vemuri,et al.  Resistance vs resilience to Alzheimer disease , 2018, Neurology.

[13]  C. Jack,et al.  NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease , 2018, Alzheimer's & Dementia.

[14]  S. DeKosky,et al.  Glymphatic system disruption as a mediator of brain trauma and chronic traumatic encephalopathy , 2018, Neuroscience & Biobehavioral Reviews.

[15]  Alzheimer's Disease Neuroimaging Initiative Association of plasma neurofilament light with neurodegeneration in patients with Alzheimer disease , 2017 .

[16]  Henrik Zetterberg,et al.  Association of Plasma Neurofilament Light With Neurodegeneration in Patients With Alzheimer Disease , 2017, JAMA neurology.

[17]  Manoj Kumar,et al.  INGE GRUNDKE-IQBAL AWARD FOR ALZHEIMER’S RESEARCH: NEUROTOXIC REACTIVE ASTROCYTES ARE INDUCED BY ACTIVATED MICROGLIA , 2019, Alzheimer's & Dementia.

[18]  Henrik Zetterberg,et al.  Neurofilament Light: A Dynamic Cross-Disease Fluid Biomarker for Neurodegeneration , 2016, Neuron.

[19]  K. Blennow,et al.  CSF and blood biomarkers for the diagnosis of Alzheimer's disease: a systematic review and meta-analysis , 2016, The Lancet Neurology.

[20]  Stephen F. Carter,et al.  Diverging longitudinal changes in astrocytosis and amyloid PET in autosomal dominant Alzheimer’s disease , 2016, Brain : a journal of neurology.

[21]  Kevin K. W. Wang,et al.  Glial fibrillary acidic protein: from intermediate filament assembly and gliosis to neurobiomarker , 2015, Trends in Neurosciences.

[22]  Maiken Nedergaard,et al.  The Glymphatic System: A Beginner’s Guide , 2015, Neurochemical Research.

[23]  O. Garaschuk,et al.  Neuroinflammation in Alzheimer's disease , 2015, The Lancet Neurology.

[24]  M. Heneka,et al.  Innate immunity in Alzheimer's disease , 2015, Nature Immunology.

[25]  Maiken Nedergaard,et al.  Biomarkers of Traumatic Injury Are Transported from Brain to Blood via the Glymphatic System , 2015, The Journal of Neuroscience.

[26]  Robert A. Koeppe,et al.  The Centiloid Project: Standardizing quantitative amyloid plaque estimation by PET , 2015, Alzheimer's & Dementia.

[27]  J. Middeldorp,et al.  Glial fibrillary acidic protein isoform expression in plaque related astrogliosis in Alzheimer's disease , 2014, Neurobiology of Aging.

[28]  K. Sharkey,et al.  Novel functional roles for enteric glia in the gastrointestinal tract , 2012, Nature Reviews Gastroenterology &Hepatology.

[29]  G. E. Vates,et al.  A Paravascular Pathway Facilitates CSF Flow Through the Brain Parenchyma and the Clearance of Interstitial Solutes, Including Amyloid β , 2012, Science Translational Medicine.

[30]  B. Barres,et al.  Genomic Analysis of Reactive Astrogliosis , 2012, The Journal of Neuroscience.

[31]  Ove Almkvist,et al.  Evidence for Astrocytosis in Prodromal Alzheimer Disease Provided by 11C-Deuterium-L-Deprenyl: A Multitracer PET Paradigm Combining 11C-Pittsburgh Compound B and 18F-FDG , 2012, The Journal of Nuclear Medicine.

[32]  Nick C Fox,et al.  The diagnosis of mild cognitive impairment due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer's disease , 2011, Alzheimer's & Dementia.

[33]  J. Morris,et al.  The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer's disease , 2011, Alzheimer's & Dementia.

[34]  N. Bogdanovic,et al.  Blood-brain barrier alterations in ageing and dementia , 2009, Journal of the Neurological Sciences.

[35]  Sid E O'Bryant,et al.  Staging dementia using Clinical Dementia Rating Scale Sum of Boxes scores: a Texas Alzheimer's research consortium study. , 2008, Archives of neurology.

[36]  N. Belyaev,et al.  Amyloid-degrading enzymes as therapeutic targets in Alzheimer's disease. , 2008, Current Alzheimer research.

[37]  Milos Pekny,et al.  Redefining the concept of reactive astrocytes as cells that remain within their unique domains upon reaction to injury , 2006, Proceedings of the National Academy of Sciences.

[38]  G. Leuba,et al.  Neuronal and Nonneuronal Quantitative BACE Immunocytochemical Expression in the Entorhinohippocampal and Frontal Regions in Alzheimer’s Disease , 2005, Dementia and Geriatric Cognitive Disorders.

[39]  T. Wyss-Coray,et al.  Adult mouse astrocytes degrade amyloid-β in vitro and in situ , 2003, Nature Network Boston.

[40]  J. Schneider,et al.  Parahippocampal tau pathology in healthy aging, mild cognitive impairment, and early Alzheimer's disease , 2002, Annals of neurology.

[41]  J. Wegiel,et al.  Microglial cells are the driving force in fibrillar plaque formation, whereas astrocytes are a leading factor in plaque degradation , 2000, Acta Neuropathologica.

[42]  H. Braak,et al.  Amyloid β-protein (Aβ)-containing astrocytes are located preferentially near N-terminal-truncated Aβ deposits in the human entorhinal cortex , 2000, Acta Neuropathologica.

[43]  M. Albert,et al.  Age at onset of Alzheimer's disease , 1994, Neurology.

[44]  J. Haines,et al.  Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. , 1993, Science.

[45]  Virginia M. Y. Lee,et al.  Neuronal modulation of schwann cell glial fibrillary acidic protein (GFAP) , 1989, Journal of neuroscience research.