1
|
Safransky M, Groh JR, Blennow K, Zetterberg H, Tripodis Y, Martin B, Weller J, Asken BM, Rabinovici GD, Qiu WWQ, McKee AC, Stein TD, Mez J, Henson RL, Long J, Morris JC, Perrin RJ, Schindler SE, Alosco ML. Lumipulse-Measured Cerebrospinal Fluid Biomarkers for the Early Detection of Alzheimer Disease. Neurology 2024; 103:e209866. [PMID: 39496102 PMCID: PMC11540457 DOI: 10.1212/wnl.0000000000209866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/20/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND AND OBJECTIVES CSF biomarkers of Aβ42 and phosphorylated tau (p-tau181) are used clinically for the detection of Alzheimer disease (AD) pathology during life. CSF biomarker validation studies have largely used clinical diagnoses and/or amyloid PET imaging as the reference standard. The few existing CSF-to-autopsy studies have been restricted to late-stage AD. This CSF-to-autopsy study investigated associations between CSF biomarkers of AD and AD neuropathologic changes among brain donors who had normal cognition at the time of lumbar puncture (LP). METHODS This was a retrospective study of brain donors from the National Alzheimer's Coordinating Center who had normal cognition at the time of LP and who had measurements of CSF Aβ42 and p-tau181 performed with Lumipulse assays. All brain donors were from Washington University Knight ADRC. Staging of AD neuropathologic change (ADNC) was made based on National Institute on Aging-Alzheimer's Association criteria. For this study, participants were divided into 2 categories: "AD-" (no AD/low ADNC) and "AD+" (intermediate/high ADNC). Accuracy of each biomarker for discriminating AD status was evaluated using area under the curve (AUC) statistics generated using predicted probabilities from binary logistic regressions that controlled for age, sex, APOE ε4, and interval between LP and death. RESULTS The average age at LP was 79.3 years (SD = 5.6), and the average age at death was 87.1 years (SD = 6.5). Of the 49 brain donors, 24 (49%) were male and 47 (95.9%) were White. 20 (40.8%) had autopsy-confirmed AD. The average interval from LP until death was 7.76 years (SD = 4.31). CSF p-tau181/Aβ42 was the optimal predictor of AD, having excellent discrimination accuracy (AUC = 0.97, 95% CI 0.94-1.00, p = 0.003). CSF p-tau181 alone had the second-best discrimination accuracy (AUC = 0.92, 95% CI 0.84-1.00, p = 0.001), followed by CSF Aβ42 alone (AUC = 0.92, 95% CI 0.85-1.00, p = 0.007), while CSF t-tau had the numerically lowest discrimination accuracy (AUC = 0.87, 95% CI 0.76-0.97, p = 0.005). Effects remained after controlling for prevalent comorbid neuropathologies. CSF p-tau181/Aβ42 was strongly associated with CERAD ratings of neuritic amyloid plaque scores and Braak staging of NFTs. DISCUSSION This study supports Lumipulse-measured CSF Aβ42 and p-tau181 and, particularly, the ratio of p-tau181 to Aβ42, for the early detection of AD pathophysiologic processes. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that Lumipulse measures of p-tau181/Aβ42 in the CSF accurately discriminated cognitively normal participants with and without Alzheimer disease neuropathologic change.
Collapse
Affiliation(s)
- Michelle Safransky
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Jenna R Groh
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Kaj Blennow
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Henrik Zetterberg
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Yorghos Tripodis
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Brett Martin
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Jason Weller
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Breton M Asken
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Gil D Rabinovici
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Wendy Wei Qiao Qiu
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Ann C McKee
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Thor D Stein
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Jesse Mez
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Rachel L Henson
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Justin Long
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - John C Morris
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Richard J Perrin
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Suzanne E Schindler
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| | - Michael L Alosco
- From the Boston University Alzheimer's Disease Research Center (M.S., J.R.G., J.W., W.W.Q.Q., A.C.M., T.D.S., J.M., M.L.A.), Boston University CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, MA; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institut du Cerveau et de la Moelle épinière (ICM) (K.B.), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France; University of Science and Technology of China and First Affiliated Hospital of USTC (K.B.), Hefei, Anhui, P.R. China; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology; UK Dementia Research Institute at UCL (H.Z.), UCL Institute of Neurology, University College London, United Kingdom; Department of Biostatistics (Y.T.); Biostatistics and Epidemiology Data Analytics Center (BEDAC) (B.M.), Boston University School of Public Health, MA; University of Florida (B.M.A.), Gainesville, FL; Memory & Aging Center (G.D.R.), Department of Neurology, Weill Institute for Neurosciences; Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Psychiatry (W.W.Q.Q.); Department of Pharmacology and Experimental Therapeutics (W.W.Q.Q.), Boston University Chobanian & Avedisian School of Medicine, MA; VA Boston Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Jamaica Plain, MA; Department of Pathology and Laboratory Medicine (A.C.M., T.D.S.), Boston University Chobanian & Avedisian School of Medicine; VA Bedford Healthcare System (A.C.M., T.D.S.), US Department of Veteran Affairs, Bedford; Framingham Heart Study (J.M.), Framingham, MA; Department of Neurology (R.L.H., J.L., J.C.M., R.J.P., S.E.S.), Knight Alzheimer's Disease Research Center, Washington University School of Medicine; Department of Neurology (M.L.A.), Boston Medical Center; and Department of Anatomy & Neurobiology (M.L.A.), Boston University Chobanian & Avedisian School of Medicine, MA
| |
Collapse
|
2
|
Behl T, Kyada A, Roopashree R, Nathiya D, Arya R, Kumar MR, Khalid M, Gulati M, Sachdeva M, Fareed M, Patra PK, Agrawal A, Wal P, Gasmi A. Epigenetic biomarkers in Alzheimer's disease: Diagnostic and prognostic relevance. Ageing Res Rev 2024; 102:102556. [PMID: 39490904 DOI: 10.1016/j.arr.2024.102556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) is a leading cause of cognitive decline in the aging population, presenting a critical need for early diagnosis and effective prognostic tools. Epigenetic modifications, including DNA methylation, histone modifications, and non-coding RNAs, have emerged as promising biomarkers for AD due to their roles in regulating gene expression and potential for reversibility. This review examines the current landscape of epigenetic biomarkers in AD, emphasizing their diagnostic and prognostic relevance. DNA methylation patterns in genes such as APP, PSEN1, and PSEN2 are highlighted for their strong associations with AD pathology. Alterations in DNA methylation at specific CpG sites have been consistently observed in AD patients, suggesting their utility in early detection. Histone modifications, such as acetylation and methylation, also play a crucial role in chromatin remodelling and gene expression regulation in AD. Dysregulated histone acetylation and methylation have been linked to AD progression, making these modifications valuable biomarkers. Non-coding RNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), further contribute to the epigenetic regulation in AD. miRNAs can modulate gene expression post-transcriptionally and have been found in altered levels in AD, while lncRNAs can influence chromatin structure and gene expression. The presence of these non-coding RNAs in biofluids like blood and cerebrospinal fluid positions them as accessible and minimally invasive biomarkers. Technological advancements in detecting and quantifying epigenetic modifications have propelled the field forward. Techniques such as next-generation sequencing, bisulfite sequencing, and chromatin immunoprecipitation assays offer high sensitivity and specificity, enabling the detailed analysis of epigenetic changes in clinical samples. These tools are instrumental in translating epigenetic research into clinical practice. This review underscores the potential of epigenetic biomarkers to enhance the early diagnosis and prognosis of AD, paving the way for personalized therapeutic strategies and improved patient outcomes. The integration of these biomarkers into clinical workflows promises to revolutionize AD management, offering hope for better disease monitoring and intervention.
Collapse
Affiliation(s)
- Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab 140306, India.
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University, Jaipur, India
| | - Renu Arya
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Mohammad Khalid
- Department of pharmacognosy, College of pharmacy, Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Monika Sachdeva
- Fatima College of Health Sciences, Al Ain, United Arab Emirates
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box No. 71666, Riyadh 11597, Saudi Arabia
| | - Pratap Kumar Patra
- School of Pharmacy & Life Sciences, Centurion University of Technology & Managemnet, Bhubaneswar, Odisha 752050, India
| | - Ankur Agrawal
- Jai Institute of Pharmaceutical Sciences and Research, Gwalior, Madhya Pradesh 474001, India
| | - Pranay Wal
- PSIT-Pranveer Singh Institute of Technology, Pharmacy, NH-19, Bhauti Road, Kanpur, UP 209305, India
| | - Amin Gasmi
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France; International Institute of Nutrition and Micronutrition Sciences, Saint-Étienne, France
| |
Collapse
|
3
|
Rezaei A, van den Berg M, Mirlohi H, Verhoye M, Amiri M, Keliris GA. Recurrence quantification analysis of rs-fMRI data: A method to detect subtle changes in the TgF344-AD rat model. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108378. [PMID: 39260164 DOI: 10.1016/j.cmpb.2024.108378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/07/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND OBJECTIVE Alzheimer's disease (AD) is one of the leading causes of dementia, affecting the world's population at a growing rate. The preclinical stage of AD lasts over a decade, hence understanding AD-related early neuropathological effects on brain function at this stage facilitates early detection of the disease. METHODS Resting-state functional magnetic resonance imaging (rs-fMRI) has been a powerful tool for understanding brain function, and it has been widely used in AD research. In this study, we apply Recurrence Quantification Analysis (RQA) on rs-fMRI images of 4-months (4 m) and 6-months-old (6 m) TgF344-AD rats and WT littermates to identify changes related to the AD phenotype and aging. RQA has been focused on areas of the default mode-like network (DMLN) and was performed based on Recurrence Plots (RP). RP is a mathematical representation of any dynamical system that evolves over time as a set of its state recurrences. In this paper, RPs were extracted in order to identify the affected regions of the DMLN at very early stages of AD. RESULTS Using the RQA approach, we identified significant changes related to the AD phenotype at 4 m and/or 6 m in several areas of the rat DMLN including the BFB, Hippocampal fields CA1 and CA3, CG1, CG2, PrL, PtA, RSC, TeA, V1, V2. In addition, with age, brain activity of WT rats showed less predictability, while the AD rats presented reduced decline of predictability. CONCLUSIONS The results of this study demonstrate that RQA of rs-fMRI data is a potent approach that can detect subtle changes which might be missed by other methodologies due to the brain's non-linear dynamics. Moreover, this study provides helpful information about specific areas involved in AD pathology at very early stages of the disease in a very promising rat model of AD. Our results provide valuable information for the development of early detection methods and novel diagnosis tools for AD.
Collapse
Affiliation(s)
- Arash Rezaei
- Medical Biology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Monica van den Berg
- Bio-Imaging Lab, University of Antwerp, Belgium; µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Hajar Mirlohi
- Medical Biology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Belgium; µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Mahmood Amiri
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Georgios A Keliris
- Bio-Imaging Lab, University of Antwerp, Belgium; µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium; Institute of Computer Science, Foundation for Research & Technology, Hellas, Heraklion, Crete, Greece
| |
Collapse
|
4
|
Ding H, Gifford K, Shih LC, Ho K, Rahman S, Igwe A, Low S, Popp Z, Searls E, Li Z, Madan S, Burk A, Hwang PH, Anda-Duran ID, Kolachalama VB, Au R, Lin H. Exploring the Perspectives of Older Adults on a Digital Brain Health Platform Using Natural Language Processing: Cohort Study. JMIR Form Res 2024; 8:e60453. [PMID: 39556805 PMCID: PMC11612578 DOI: 10.2196/60453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Although digital technology represents a growing field aiming to revolutionize early Alzheimer disease risk prediction and monitoring, the perspectives of older adults on an integrated digital brain health platform have not been investigated. OBJECTIVE This study aims to understand the perspectives of older adults on a digital brain health platform by conducting semistructured interviews and analyzing their transcriptions by natural language processing. METHODS The study included 28 participants from the Boston University Alzheimer's Disease Research Center, all of whom engaged with a digital brain health platform over an initial assessment period of 14 days. Semistructured interviews were conducted to collect data on participants' experiences with the digital brain health platform. The transcripts generated from these interviews were analyzed using natural language processing techniques. The frequency of positive and negative terms was evaluated through word count analysis. A sentiment analysis was used to measure the emotional tone and subjective perceptions of the participants toward the digital platform. RESULTS Word count analysis revealed a generally positive sentiment toward the digital platform, with "like," "well," and "good" being the most frequently mentioned positive terms. However, terms such as "problem" and "hard" indicated certain challenges faced by participants. Sentiment analysis showed a slightly positive attitude with a median polarity score of 0.13 (IQR 0.08-0.15), ranging from -1 (completely negative) to 1 (completely positive), and a median subjectivity score of 0.51 (IQR 0.47-0.53), ranging from 0 (completely objective) to 1 (completely subjective). These results suggested an overall positive attitude among the study cohort. CONCLUSIONS The study highlights the importance of understanding older adults' attitudes toward digital health platforms amid the comprehensive evolution of the digitalization era. Future research should focus on refining digital solutions to meet the specific needs of older adults, fostering a more personalized approach to brain health.
Collapse
Affiliation(s)
- Huitong Ding
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- The Framingham Heart Study, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Katherine Gifford
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- The Framingham Heart Study, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Ludy C Shih
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Kristi Ho
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Salman Rahman
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Akwaugo Igwe
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Spencer Low
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Zachary Popp
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Edward Searls
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Zexu Li
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Sanskruti Madan
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Alexa Burk
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Phillip H Hwang
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- The Framingham Heart Study, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Ileana De Anda-Duran
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Vijaya B Kolachalama
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- Department of Computer Science, Boston University, Boston, MA, United States
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- The Framingham Heart Study, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
- Department of Medicine, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- Slone Epidemiology Center, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
- Departments of Neurology, Avedisian School of Medicine, Boston University Chobanian, Boston, MA, United States
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
5
|
Bigler ED, Allder S, Victoroff J. What traditional neuropsychological assessment got wrong about mild traumatic brain injury. II: limitations in test development, research design, statistical and psychometric issues. Brain Inj 2024; 38:1053-1074. [PMID: 39066740 DOI: 10.1080/02699052.2024.2376261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/16/2024] [Accepted: 06/30/2024] [Indexed: 07/30/2024]
Abstract
PRIMARY OBJECTIVE This is Part II of a four-part opinion review on traditional neuropsychological assessment methods and findings associated with mild traumatic brain injury (mTBI). This Part II review focuses on historical, psychometric and statistical issues involving traditional neuropsychological methods that have been used in neuropsychological outcome studies of mTBI, but demonstrates the critical limitations of traditional methods. RESEARCH DESIGN This is an opinion review. METHODS AND PROCEDURES Traditional neuropsychological tests are dated and lack specificity in evaluating such a heterogenous and complex injury as occurs with mTBI. MAIN OUTCOME AND RESULTS In this review, we demonstrate traditional neuropsychological methods were never developed as standalone measures for detecting subtle changes in neurocognitive or neurobehavioral functioning and likewise, never designed to address the multifaceted issues related to underlying mTBI neuropathology symptom burden from having sustained a concussive brain injury. CONCLUSIONS For neuropsychological assessment to continue to contribute to clinical practice and outcome literature involving mTBI, major innovative changes are needed that will likely require technological advances of novel assessment techniques more specifically directed to evaluating the mTBI patient. These will be discussed in Part IV.
Collapse
Affiliation(s)
- Erin D Bigler
- Department of Psychology and Neuroscience Center, Brigham Young University, Provo, Utah, USA
- Departments of Neurology and Psychiatry, University of Utah, Salt Lake City, Utah, USA
| | - Steven Allder
- Consultant Neurologist and Clinical Director, Re: Cognition Health, London, UK
| | - Jeff Victoroff
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
He B, Wu R, Sangani N, Pugalenthi PV, Patania A, Risacher SL, Nho K, Apostolova LG, Shen L, Saykin AJ, Yan J. Integrating amyloid imaging and genetics for early risk stratification of Alzheimer's disease. Alzheimers Dement 2024; 20:7819-7830. [PMID: 39285750 PMCID: PMC11567859 DOI: 10.1002/alz.14244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/24/2024] [Accepted: 08/15/2024] [Indexed: 09/21/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) initiates years prior to symptoms, underscoring the importance of early detection. While amyloid accumulation starts early, individuals with substantial amyloid burden may remain cognitively normal, implying that amyloid alone is not sufficient for early risk assessment. METHODS Given the genetic susceptibility of AD, a multi-factorial pseudotime approach was proposed to integrate amyloid imaging and genotype data for estimating a risk score. Validation involved association with cognitive decline and survival analysis across risk-stratified groups, focusing on patients with mild cognitive impairment (MCI). RESULTS Our risk score outperformed amyloid composite standardized uptake value ratio in correlation with cognitive scores. MCI subjects with lower pseudotime risk score showed substantial delayed onset of AD and slower cognitive decline. Moreover, pseudotime risk score demonstrated strong capability in risk stratification within traditionally defined subgroups such as early MCI, apolipoprotein E (APOE) ε4+ MCI, APOE ε4- MCI, and amyloid+ MCI. DISCUSSION Our risk score holds great potential to improve the precision of early risk assessment. HIGHLIGHTS Accurate early risk assessment is critical for the success of clinical trials. A new risk score was built from integrating amyloid imaging and genetic data. Our risk score demonstrated improved capability in early risk stratification.
Collapse
Affiliation(s)
- Bing He
- Department of Biomedical Engineering and InformaticsIndiana University Luddy School of Informatics, Computing and EngineeringIndianapolisIndianaUSA
| | - Ruiming Wu
- Department of Biomedical Engineering and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Neel Sangani
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Pradeep Varathan Pugalenthi
- Department of Biomedical Engineering and InformaticsIndiana University Luddy School of Informatics, Computing and EngineeringIndianapolisIndianaUSA
| | - Alice Patania
- Department of Mathematics StatisticsUniversity of VermontBurlingtonVermontUSA
| | - Shannon L. Risacher
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kwangsik Nho
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Liana G. Apostolova
- Department of Biomedical Engineering and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Li Shen
- Department of Biomedical Engineering and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Andrew J. Saykin
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jingwen Yan
- Department of Biomedical Engineering and InformaticsIndiana University Luddy School of Informatics, Computing and EngineeringIndianapolisIndianaUSA
| | | |
Collapse
|
7
|
Doke R, Lamkhade GJ, Vinchurkar K, Singh S. Demystifying the Role of Neuroinflammatory Mediators as Biomarkers for Diagnosis, Prognosis, and Treatment of Alzheimer's Disease: A Review. ACS Pharmacol Transl Sci 2024; 7:2987-3003. [PMID: 39416969 PMCID: PMC11475310 DOI: 10.1021/acsptsci.4c00457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Neuroinflammatory mediators play a pivotal role in the pathogenesis of Alzheimer's Disease (AD), influencing its onset, progression, and severity. The precise mechanisms behind AD are still not fully understood, leading current treatments to focus mainly on managing symptoms rather than preventing or curing the condition. The amyloid and tau hypotheses are the most widely accepted explanations for AD pathology; however, they do not completely account for the neuronal degeneration observed in AD. Growing evidence underscores the crucial role of neuroinflammation in the pathology of AD. The neuroinflammatory hypothesis presents a promising new approach to understanding the mechanisms driving AD. This review examines the importance of neuroinflammatory biomarkers in the diagnosis, prognosis, and treatment of AD. It delves into the mechanisms underlying neuroinflammation in AD, highlighting the involvement of various mediators such as cytokines, chemokines, and ROS. Additionally, this review discusses the potential of neuroinflammatory biomarkers as diagnostic tools, prognostic indicators, and therapeutic targets for AD management. By understanding the intricate interplay between neuroinflammation and AD pathology, this review aims to help in the development of efficient diagnostic and treatment plans to fight this debilitating neurological condition. Furthermore, it elaborates recent advancements in neuroimaging techniques and biofluid analysis for the identification and monitoring of neuroinflammatory biomarkers in AD patients.
Collapse
Affiliation(s)
- Rohit
R. Doke
- Jaihind
College of Pharmacy, Vadgaon Sahani, Pune, Maharashtra 412401, India
| | | | - Kuldeep Vinchurkar
- Krishna
School of Pharmacy, Kiran and Pallavi Patel
Global University, Vadodara, Gujarat 391243, India
| | - Sudarshan Singh
- Office
of Research Administration, Chiang Mai University, Chaing Mai 50200, Thailand
- Faculty
of Pharmacy, Chiang Mai University, Chaing Mai 50200, Thailand
| |
Collapse
|
8
|
Smith RG, Pishva E, Kouhsar M, Imm J, Dobricic V, Johannsen P, Wittig M, Franke A, Vandenberghe R, Schaeverbeke J, Freund‐Levi Y, Frölich L, Scheltens P, Teunissen CE, Frisoni G, Blin O, Richardson JC, Bordet R, Engelborghs S, de Roeck E, Martinez‐Lage P, Altuna M, Tainta M, Lleó A, Sala I, Popp J, Peyratout G, Winchester L, Nevado‐Holgado A, Verhey F, Tsolaki M, Andreasson U, Blennow K, Zetterberg H, Streffer J, Vos SJB, Lovestone S, Visser PJ, Bertram L, Lunnon K. Blood DNA methylomic signatures associated with CSF biomarkers of Alzheimer's disease in the EMIF-AD study. Alzheimers Dement 2024; 20:6722-6739. [PMID: 39193893 PMCID: PMC11485320 DOI: 10.1002/alz.14098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION We investigated blood DNA methylation patterns associated with 15 well-established cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) pathophysiology, neuroinflammation, and neurodegeneration. METHODS We assessed DNA methylation in 885 blood samples from the European Medical Information Framework for Alzheimer's Disease (EMIF-AD) study using the EPIC array. RESULTS We identified Bonferroni-significant differential methylation associated with CSF YKL-40 (five loci) and neurofilament light chain (NfL; seven loci) levels, with two of the loci associated with CSF YKL-40 levels correlating with plasma YKL-40 levels. A co-localization analysis showed shared genetic variants underlying YKL-40 DNA methylation and CSF protein levels, with evidence that DNA methylation mediates the association between genotype and protein levels. Weighted gene correlation network analysis identified two modules of co-methylated loci correlated with several amyloid measures and enriched in pathways associated with lipoproteins and development. DISCUSSION We conducted the most comprehensive epigenome-wide association study (EWAS) of AD-relevant CSF biomarkers to date. Future work should explore the relationship between YKL-40 genotype, DNA methylation, and protein levels in the brain. HIGHLIGHTS Blood DNA methylation was assessed in the EMIF-AD MBD study. Epigenome-wide association studies (EWASs) were performed for 15 Alzheimer's disease (AD)-relevant cerebrospinal fluid (CSF) biomarker measures. Five Bonferroni-significant loci were associated with YKL-40 levels and seven with neurofilament light chain (NfL). DNA methylation in YKL-40 co-localized with previously reported genetic variation. DNA methylation potentially mediates the effect of single-nucleotide polymorphisms (SNPs) in YKL-40 on CSF protein levels.
Collapse
Affiliation(s)
- Rebecca G. Smith
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Ehsan Pishva
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
| | - Morteza Kouhsar
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Jennifer Imm
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA)University of LübeckLübeckGermany
| | - Peter Johannsen
- Danish Dementia Research Centre, RigshospitaletCopenhagenDenmark
| | - Michael Wittig
- Institute of Clinical Molecular BiologyChristian‐Albrechts‐University of KielKielGermany
| | - Andre Franke
- Institute of Clinical Molecular BiologyChristian‐Albrechts‐University of KielKielGermany
| | - Rik Vandenberghe
- Laboratory for Cognitive NeurologyKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | - Jolien Schaeverbeke
- Laboratory for Cognitive NeurologyKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | - Yvonne Freund‐Levi
- Department of Clinical Science and EducationSödersjukhuset, Karolinska InstitutetStockholmSweden
- School of Medical SciencesÖrebro UniversityÖrebroSweden
- Department of GeriatricsSödertälje HospitalSödertäljeSweden
| | - Lutz Frölich
- Department of Geriatric PsychiatryCentral Institut of Mental HealthMedical Faculty Mannheim/Heidelberg UniversityMannheimGermany
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam NeuroscienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Laboratory Medicine, Amsterdam NeuroscienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Giovanni Frisoni
- Memory centerGeneva University and University Hospitals; on behalf of the AMYPAD consortiumGenevaSwitzerland
| | | | - Jill C. Richardson
- Neuroscience Therapeutic Area, GlaxoSmithKline R&DStevenageHertfordshireUK
| | | | - Sebastiaan Engelborghs
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N)Vrije Universiteit Brussel (VUB), JetteBrusselsBelgium
| | - Ellen de Roeck
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | - Pablo Martinez‐Lage
- Center for Research and Advanced TherapiesFundación CITA‐Alzhéimer FundazioaSan SebastianGipuzkoaSpain
| | - Miren Altuna
- Center for Research and Advanced TherapiesFundación CITA‐Alzhéimer FundazioaSan SebastianGipuzkoaSpain
| | - Mikel Tainta
- Center for Research and Advanced TherapiesFundación CITA‐Alzhéimer FundazioaSan SebastianGipuzkoaSpain
| | - Alberto Lleó
- Servicio de Neurología, Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)Hospital Sant PauBarcelonaSpain
| | - Isabel Sala
- Servicio de Neurología, Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)Hospital Sant PauBarcelonaSpain
| | - Julius Popp
- University Hospital of Psychiatry Zürich, University of ZürichZürichSwitzerland
| | - Gwendoline Peyratout
- Department of PsychiatryUniversity Hospital of Lausanne (CHUV)LausanneSwitzerland
| | | | | | - Frans Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
| | - Magda Tsolaki
- 1st Department of NeurologySchool of MedicineLaboratory of Neurodegenerative DiseasesCenter for Interdisciplinary Research and InnovationAristotle University of Thessaloniki, and Alzheimer HellasThessalonikiGreece
| | - Ulf Andreasson
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
| | - Kaj Blennow
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
- Paris Brain InstituteICM, Pitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiPR China
| | - Henrik Zetterberg
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative Diseases, N.T.ShatinHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public Health, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | | | - Stephanie J. B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
| | - Simon Lovestone
- Department of PsychiatryUniversity Hospital of Lausanne (CHUV)LausanneSwitzerland
- Currently at: Johnson & Johnson Innovative MedicinesBeerseBelgium
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam NeuroscienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA)University of LübeckLübeckGermany
| | - Katie Lunnon
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| |
Collapse
|
9
|
Panizza E, Cerione RA. An interpretable deep learning framework identifies proteomic drivers of Alzheimer's disease. Front Cell Dev Biol 2024; 12:1379984. [PMID: 39355118 PMCID: PMC11442384 DOI: 10.3389/fcell.2024.1379984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/22/2024] [Indexed: 10/03/2024] Open
Abstract
Alzheimer's disease (AD) is the leading neurodegenerative pathology in aged individuals, but many questions remain on its pathogenesis, and a cure is still not available. Recent research efforts have generated measurements of multiple omics in individuals that were healthy or diagnosed with AD. Although machine learning approaches are well-suited to handle the complexity of omics data, the models typically lack interpretability. Additionally, while the genetic landscape of AD is somewhat more established, the proteomic landscape of the diseased brain is less well-understood. Here, we establish a deep learning method that takes advantage of an ensemble of autoencoders (AEs) - EnsembleOmicsAE-to reduce the complexity of proteomics data into a reduced space containing a small number of latent features. We combine brain proteomic data from 559 individuals across three AD cohorts and demonstrate that the ensemble autoencoder models generate stable latent features which are well-suited for downstream biological interpretation. We present an algorithm to calculate feature importance scores based on the iterative scrambling of individual input features (i.e., proteins) and show that the algorithm identifies signaling modules (AE signaling modules) that are significantly enriched in protein-protein interactions. The molecular drivers of AD identified within the AE signaling modules derived with EnsembleOmicsAE were missed by linear methods, including integrin signaling and cell adhesion. Finally, we characterize the relationship between the AE signaling modules and the age of death of the patients and identify a differential regulation of vimentin and MAPK signaling in younger compared with older AD patients.
Collapse
Affiliation(s)
- Elena Panizza
- Department of Molecular Medicine, Cornell University, Ithaca, NY, United States
| | - Richard A. Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, NY, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, United States
| |
Collapse
|
10
|
Bader I, Groot C, Tan HS, Milongo JMA, Haan JD, Verberk IMW, Yong K, Orellina J, Campbell S, Wilson D, van Harten AC, Kok PHB, van der Flier WM, Pijnenburg YAL, Barkhof F, van de Giessen E, Teunissen CE, Bouwman FH, Ossenkoppele R. Rationale and design of the BeyeOMARKER study: prospective evaluation of blood- and eye-based biomarkers for early detection of Alzheimer's disease pathology in the eye clinic. Alzheimers Res Ther 2024; 16:190. [PMID: 39169442 PMCID: PMC11340081 DOI: 10.1186/s13195-024-01545-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a common, complex and multifactorial disease that may require screening across multiple routes of referral to enable early detection and subsequent future implementation of tailored interventions. Blood- and eye-based biomarkers show promise as low-cost, scalable and patient-friendly tools for early AD detection given their ability to provide information on AD pathophysiological changes and manifestations in the retina, respectively. Eye clinics provide an intriguing real-world proof-of-concept setting to evaluate the performance of these potential AD screening tools given the intricate connections between the eye and brain, presumed enrichment for AD pathology in the aging population with eye disorders, and the potential for an accelerated diagnostic pathway for under-recognized patient groups. METHODS The BeyeOMARKER study is a prospective, observational, longitudinal cohort study aiming to include individuals visiting an eye-clinic. Inclusion criteria entail being ≥ 50 years old and having no prior dementia diagnosis. Excluded eye-conditions include traumatic insults, superficial inflammation, and conditions in surrounding structures of the eye that are not engaged in vision. The BeyeOMARKER cohort (n = 700) will undergo blood collection to assess plasma p-tau217 levels and a brief cognitive screening at the eye clinic. All participants will subsequently be invited for annual longitudinal follow-up including remotely administered cognitive screening and questionnaires. The BeyeOMARKER + cohort (n = 150), consisting of 100 plasma p-tau217 positive participants and 50 matched negative controls selected from the BeyeOMARKER cohort, will additionally undergo Aβ-PET and tau-PET, MRI, retinal imaging including hyperspectral imaging (primary), widefield imaging, optical coherence tomography (OCT) and OCT-Angiography (secondary), and cognitive and cortical vision assessments. RESULTS We aim to implement the current protocol between April 2024 until March 2027. Primary outcomes include the performance of plasma p-tau217 and hyperspectral retinal imaging to detect AD pathology (using Aβ- and tau-PET visual read as reference standard) and to detect cognitive decline. Initial follow-up is ~ 2 years but may be extended with additional funding. CONCLUSIONS We envision that the BeyeOMARKER study will demonstrate the feasibility of early AD detection based on blood- and eye-based biomarkers in alternative screening settings, and will improve our understanding of the eye-brain connection. TRIAL REGISTRATION The BeyeOMARKER study (Eudamed CIV ID: CIV-NL-23-09-044086; registration date: 19th of March 2024) is approved by the ethical review board of the Amsterdam UMC.
Collapse
Affiliation(s)
- Ilse Bader
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands.
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands.
| | - Colin Groot
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - H Stevie Tan
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, 1081 HV, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Amsterdam UMC Location VUmc, Amsterdam Reproduction and Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Jean-Marie A Milongo
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
| | - Jurre den Haan
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Laboratory Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Keir Yong
- Queen Square Institute of Neurology, Dementia Research Centre, London, WC1N 3BG, UK
| | | | | | | | - Argonde C van Harten
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Pauline H B Kok
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
| | - Wiesje M van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Yolande A L Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Frederik Barkhof
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HZ, The Netherlands
- UCL Queen Square Institute of Neurology and Centre for Medical Image Computing, University College, London, WC1N 3BG, UK
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Neurochemistry Laboratory, Laboratory Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Femke H Bouwman
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Rik Ossenkoppele
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands.
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
| |
Collapse
|
11
|
Yoon JH, Lee H, Kwon D, Lee D, Lee S, Cho E, Kim J, Kim D. Integrative approach of omics and imaging data to discover new insights for understanding brain diseases. Brain Commun 2024; 6:fcae265. [PMID: 39165479 PMCID: PMC11334939 DOI: 10.1093/braincomms/fcae265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/03/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024] Open
Abstract
Treatments that can completely resolve brain diseases have yet to be discovered. Omics is a novel technology that allows researchers to understand the molecular pathways underlying brain diseases. Multiple omics, including genomics, transcriptomics and proteomics, and brain imaging technologies, such as MRI, PET and EEG, have contributed to brain disease-related therapeutic target detection. However, new treatment discovery remains challenging. We focused on establishing brain multi-molecular maps using an integrative approach of omics and imaging to provide insights into brain disease diagnosis and treatment. This approach requires precise data collection using omics and imaging technologies, data processing and normalization. Incorporating a brain molecular map with the advanced technologies through artificial intelligence will help establish a system for brain disease diagnosis and treatment through regulation at the molecular level.
Collapse
Affiliation(s)
- Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Hagyeong Lee
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Dayoung Kwon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Dongha Lee
- Cognitive Science Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Seulah Lee
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Eunji Cho
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Jaehoon Kim
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Dayea Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu 41061, Republic of Korea
| |
Collapse
|
12
|
Assfaw AD, Schindler SE, Morris JC. Advances in blood biomarkers for Alzheimer disease (AD): A review. Kaohsiung J Med Sci 2024; 40:692-698. [PMID: 38888066 DOI: 10.1002/kjm2.12870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Alzheimer disease (AD) and Alzheimer Disease and Related Dementias (AD/ADRD) are growing public health challenges globally affecting millions of older adults, necessitating concerted efforts to advance our understanding and management of these conditions. AD is a progressive neurodegenerative disorder characterized pathologically by amyloid plaques and tau neurofibrillary tangles that are the primary cause of dementia in older individuals. Early and accurate diagnosis of AD dementia is crucial for effective intervention and treatment but has proven challenging to accomplish. Although testing for AD brain pathology with cerebrospinal fluid (CSF) or positron emission tomography (PET) has been available for over 2 decades, most patients never underwent this testing because of inaccessibility, high out-of-pocket costs, perceived risks, and the lack of AD-specific treatments. However, in recent years, rapid progress has been made in developing blood biomarkers for AD/ADRD. Consequently, blood biomarkers have emerged as promising tools for non-invasive and cost-effective diagnosis, prognosis, and monitoring of AD progression. This review presents the evolving landscape of blood biomarkers in AD/ADRD and explores their potential applications in clinical practice for early detection, prognosis, and therapeutic interventions. It covers recent advances in blood biomarkers, including amyloid beta (Aβ) peptides, tau protein, neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP). It also discusses their diagnostic and prognostic utility while addressing associated challenges and limitations. Future research directions in this rapidly evolving field are also proposed.
Collapse
Affiliation(s)
- Araya Dimtsu Assfaw
- Department of Neurology, Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne E Schindler
- Department of Neurology, Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, Missouri, USA
| | - John C Morris
- Department of Neurology, Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
13
|
Ma ZL, Wang ZL, Zhang FY, Liu HX, Mao LH, Yuan L. Biomarkers of Parkinson's Disease: From Basic Research to Clinical Practice. Aging Dis 2024; 15:1813-1830. [PMID: 37815899 PMCID: PMC11272192 DOI: 10.14336/ad.2023.1005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized pathologically by dopaminergic neuron loss and the formation of Lewy bodies, which are enriched with aggregated α-synuclein (α-syn). PD currently has no cure, but therapeutic strategies are available to alleviate symptoms. Early diagnosis can greatly improve therapeutic interventions, but the clinical diagnosis of PD remains challenging and depends mainly on clinical features and imaging tests. Efficient and specific biomarkers are crucial for the diagnosis, monitoring, and evaluation of PD. Here, we reviewed the biomarkers of PD in different tissues and biofluids, along with the current clinical biochemical detection methods. We found that the sensitivity and specificity of single biomarkers are limited, and selecting appropriate indicators for combined detection can improve the diagnostic accuracy of PD.
Collapse
Affiliation(s)
| | | | - Fei-yue Zhang
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Hong-xun Liu
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Li-hong Mao
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Lin Yuan
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| |
Collapse
|
14
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Sun M, Chen Z. Unveiling the Complex Role of Exosomes in Alzheimer's Disease. J Inflamm Res 2024; 17:3921-3948. [PMID: 38911990 PMCID: PMC11193473 DOI: 10.2147/jir.s466821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative illness, characterized by memory loss and cognitive decline, accounting for 60-80% of dementia cases. AD is characterized by senile plaques made up of amyloid β (Aβ) protein, intracellular neurofibrillary tangles caused by hyperphosphorylation of tau protein linked with microtubules, and neuronal loss. Currently, therapeutic treatments and nanotechnological developments are effective in treating the symptoms of AD, but a cure for the illness has not yet been found. Recently, the increased study of extracellular vesicles (EVs) has led to a growing awareness of their significant involvement in neurodegenerative disorders, including AD. Exosomes are small extracellular vesicles that transport various components including messenger RNAs, non-coding RNAs, proteins, lipids, DNA, and other bioactive compounds from one cell to another, facilitating information transmission and material movement. There is growing evidence indicating that exosomes have complex functions in AD. Exosomes may have a dual role in Alzheimer's disease by contributing to neuronal death and also helping to alleviate the pathological progression of the disease. Therefore, the primary aim of this review is to outline the updated understandings on exosomes biogenesis and many functions of exosomes in the generation, conveyance, distribution, and elimination of hazardous proteins related to Alzheimer's disease. This review is intended to provide novel insights for understanding the development, specific treatment, and early detection of Alzheimer's disease.
Collapse
Affiliation(s)
- Mingyue Sun
- Department of Neurology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Zhuoyou Chen
- Department of Neurology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| |
Collapse
|
16
|
Yang X, Sullivan PF, Li B, Fan Z, Ding D, Shu J, Guo Y, Paschou P, Bao J, Shen L, Ritchie MD, Nave G, Platt ML, Li T, Zhu H, Zhao B. Multi-organ imaging-derived polygenic indexes for brain and body health. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.04.18.23288769. [PMID: 38883759 PMCID: PMC11177904 DOI: 10.1101/2023.04.18.23288769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The UK Biobank (UKB) imaging project is a crucial resource for biomedical research, but is limited to 100,000 participants due to cost and accessibility barriers. Here we used genetic data to predict heritable imaging-derived phenotypes (IDPs) for a larger cohort. We developed and evaluated 4,375 IDP genetic scores (IGS) derived from UKB brain and body images. When applied to UKB participants who were not imaged, IGS revealed links to numerous phenotypes and stratified participants at increased risk for both brain and somatic diseases. For example, IGS identified individuals at higher risk for Alzheimer's disease and multiple sclerosis, offering additional insights beyond traditional polygenic risk scores of these diseases. When applied to independent external cohorts, IGS also stratified those at high disease risk in the All of Us Research Program and the Alzheimer's Disease Neuroimaging Initiative study. Our results demonstrate that, while the UKB imaging cohort is largely healthy and may not be the most enriched for disease risk management, it holds immense potential for stratifying the risk of various brain and body diseases in broader external genetic cohorts.
Collapse
Affiliation(s)
- Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxuan Li
- UCLA Samueli School of Engineering, Los Angeles, CA 90095, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dezheng Ding
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Juan Shu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yuxin Guo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gideon Nave
- Marketing Department, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael L. Platt
- Marketing Department, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Applied Mathematics and Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
Shang NY, Huang LJ, Lan JQ, Kang YY, Tang JS, Wang HY, Li XN, Sun Z, Chen QY, Liu MY, Wen ZP, Feng XH, Wu L, Peng Y. PHPB ameliorates memory deficits and reduces oxidative injury in Alzheimer's disease mouse model by activating Nrf2 signaling pathway. Acta Pharmacol Sin 2024; 45:1142-1159. [PMID: 38409216 PMCID: PMC11130211 DOI: 10.1038/s41401-024-01240-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/06/2024] [Indexed: 02/28/2024] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is the most common cause of dementia in elderly people and substantially affects patient quality of life. Oxidative stress is considered a key factor in the development of AD. Nrf2 plays a vital role in maintaining redox homeostasis and regulating neuroinflammatory responses in AD. Previous studies show that potassium 2-(1-hydroxypentyl)-benzoate (PHPB) exerts neuroprotective effects against cognitive impairment in a variety of dementia animal models such as APP/PS1 transgenic mice. In this study we investigated whether PHPB ameriorated the progression of AD by reducing oxidative stress (OS) damage. Both 5- and 13-month-old APP/PS1 mice were administered PHPB (100 mg·kg-1·d-1, i.g.) for 10 weeks. After the cognition assessment, the mice were euthanized, and the left hemisphere of the brain was harvested for analyses. We showed that 5-month-old APP/PS1 mice already exhibited impaired performance in the step-down test, and knockdown of Nrf2 gene only slightly increased the impairment, while knockdown of Nrf2 gene in 13-month-old APP/PS1 mice resulted in greatly worse performance. PHPB administration significantly ameliorated the cognition impairments and enhanced antioxidative capacity in APP/PS1 mice. In addition, PHPB administration significantly increased the p-AKT/AKT and p-GSK3β/GSK3β ratios and the expression levels of Nrf2, HO-1 and NQO-1 in APP/PS1 mice, but these changes were abolished by knockdown of Nrf2 gene. In SK-N-SH APPwt cells and primary mouse neurons, PHPB (10 μM) significantly increased the p-AKT/AKT and p-GSK3β/GSK3β ratios and the level of Nrf2, which were blocked by knockdown of Nrf2 gene. In summary, this study demonstrates that PHPB exerts a protective effect via the Akt/GSK3β/Nrf2 pathway and it might be a promising neuroprotective agent for the treatment of AD.
Collapse
Affiliation(s)
- Nian-Ying Shang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Long-Jian Huang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jia-Qi Lan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yu-Ying Kang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jing-Shu Tang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Hong-Yue Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xin-Nan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Zhuo Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Qiu-Yu Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Meng-Yao Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Zi-Peng Wen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xin-Hong Feng
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Ying Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
18
|
Maru K, Singh A, Jangir R, Jangir KK. Amyloid detection in neurodegenerative diseases using MOFs. J Mater Chem B 2024; 12:4553-4573. [PMID: 38646795 DOI: 10.1039/d4tb00373j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Neurodegenerative diseases (amyloid diseases such as Alzheimer's and Parkinson's), stemming from protein misfolding and aggregation, encompass a spectrum of disorders with severe systemic implications. Timely detection is pivotal in managing these diseases owing to their significant impact on organ function and high mortality rates. The diverse array of amyloid disorders, spanning localized and systemic manifestations, underscores the complexity of these conditions and highlights the need for advanced detection methods. Traditional approaches have focused on identifying biomarkers using imaging techniques (PET and MRI) or invasive procedures. However, recent efforts have focused on the use of metal-organic frameworks (MOFs), a versatile class of materials known for their unique properties, in revolutionizing amyloid disease detection. The high porosity, customizable structures, and biocompatibility of MOFs enable their integration with biomolecules, laying the groundwork for highly sensitive and specific biosensors. These sensors have been employed using electrochemical and photophysical techniques that target amyloid species under neurodegenerative conditions. The adaptability of MOFs allows for the precise detection and quantification of amyloid proteins, offering potential advancements in early diagnosis and disease management. This review article delves into how MOFs contribute to detecting amyloid diseases by categorizing their uses based on different sensing methods, such as electrochemical (EC), electrochemiluminescence (ECL), fluorescence, Förster resonance energy transfer (FRET), up-conversion luminescence resonance energy transfer (ULRET), and photoelectrochemical (PEC) sensing. The drawbacks of MOF biosensors and the challenges encountered in the field are also briefly explored from our perspective.
Collapse
Affiliation(s)
- Ketan Maru
- Sardar Vallabhbhai National Institute of Technology, Ichchanath, Surat-395 007, Gujarat, India.
| | - Amarendra Singh
- Sardar Vallabhbhai National Institute of Technology, Ichchanath, Surat-395 007, Gujarat, India.
| | - Ritambhara Jangir
- Sardar Vallabhbhai National Institute of Technology, Ichchanath, Surat-395 007, Gujarat, India.
| | | |
Collapse
|
19
|
Chen A, Shea D, Daggett V. Performance of SOBA-AD blood test in discriminating Alzheimer's disease patients from cognitively unimpaired controls in two independent cohorts. Sci Rep 2024; 14:7946. [PMID: 38575622 PMCID: PMC10995183 DOI: 10.1038/s41598-024-57107-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
Amyloid-beta (Aβ) toxic oligomers are critical early players in the molecular pathology of Alzheimer's disease (AD). We have developed a Soluble Oligomer Binding Assay (SOBA-AD) for detection of these Aβ oligomers that contain α-sheet secondary structure that discriminates plasma samples from patients on the AD continuum from non-AD controls. We tested 265 plasma samples from two independent cohorts to investigate the performance of SOBA-AD. Testing was performed at two different sites, with different personnel, reagents, and instrumentation. Across two cohorts, SOBA-AD discriminated AD patients from cognitively unimpaired (CU) subjects with 100% sensitivity, > 95% specificity, and > 98% area under the curve (AUC) (95% CI 0.95-1.00). A SOBA-AD positive readout, reflecting α-sheet toxic oligomer burden, was found in AD patients, and not in controls, providing separation of the two populations, aside from 5 SOBA-AD positive controls. Based on an earlier SOBA-AD study, the Aβ oligomers detected in these CU subjects may represent preclinical cases of AD. The results presented here support the value of SOBA-AD as a promising blood-based tool for the detection and confirmation of AD.
Collapse
Affiliation(s)
- Amy Chen
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA
| | - Dylan Shea
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA
- University of Washington, Box 355610, Seattle, WA, 98195-5610, USA
| | - Valerie Daggett
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA.
- University of Washington, Box 355610, Seattle, WA, 98195-5610, USA.
| |
Collapse
|
20
|
Jannati A, Toro-Serey C, Gomes-Osman J, Banks R, Ciesla M, Showalter J, Bates D, Tobyne S, Pascual-Leone A. Digital Clock and Recall is superior to the Mini-Mental State Examination for the detection of mild cognitive impairment and mild dementia. Alzheimers Res Ther 2024; 16:2. [PMID: 38167251 PMCID: PMC10759368 DOI: 10.1186/s13195-023-01367-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Disease-modifying treatments for Alzheimer's disease highlight the need for early detection of cognitive decline. However, at present, most primary care providers do not perform routine cognitive testing, in part due to a lack of access to practical cognitive assessments, as well as time and resources to administer and interpret the tests. Brief and sensitive digital cognitive assessments, such as the Digital Clock and Recall (DCR™), have the potential to address this need. Here, we examine the advantages of DCR over the Mini-Mental State Examination (MMSE) in detecting mild cognitive impairment (MCI) and mild dementia. METHODS We studied 706 participants from the multisite Bio-Hermes study (age mean ± SD = 71.5 ± 6.7; 58.9% female; years of education mean ± SD = 15.4 ± 2.7; primary language English), classified as cognitively unimpaired (CU; n = 360), mild cognitive impairment (MCI; n = 234), or probable mild Alzheimer's dementia (pAD; n = 111) based on a review of medical history with selected cognitive and imaging tests. We evaluated cognitive classifications (MCI and early dementia) based on the DCR and the MMSE against cohorts based on the results of the Rey Auditory Verbal Learning Test (RAVLT), the Trail Making Test-Part B (TMT-B), and the Functional Activities Questionnaire (FAQ). We also compared the influence of demographic variables such as race (White vs. Non-White), ethnicity (Hispanic vs. Non-Hispanic), and level of education (≥ 15 years vs. < 15 years) on the DCR and MMSE scores. RESULTS The DCR was superior on average to the MMSE in classifying mild cognitive impairment and early dementia, AUC = 0.70 for the DCR vs. 0.63 for the MMSE. DCR administration was also significantly faster (completed in less than 3 min regardless of cognitive status and age). Among 104 individuals who were labeled as "cognitively unimpaired" by the MMSE (score ≥ 28) but actually had verbal memory impairment as confirmed by the RAVLT, the DCR identified 84 (80.7%) as impaired. Moreover, the DCR score was significantly less biased by ethnicity than the MMSE, with no significant difference in the DCR score between Hispanic and non-Hispanic individuals. CONCLUSIONS DCR outperforms the MMSE in detecting and classifying cognitive impairment-in a fraction of the time-while being not influenced by a patient's ethnicity. The results support the utility of DCR as a sensitive and efficient cognitive assessment in primary care settings. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT04733989.
Collapse
Affiliation(s)
- Ali Jannati
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
| | - Claudio Toro-Serey
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA
| | - Joyce Gomes-Osman
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Russell Banks
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA
- Department of Communicative Sciences & Disorders, Michigan State University, East Lansing, MI, USA
| | - Marissa Ciesla
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA
| | - John Showalter
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA
| | - David Bates
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA
| | - Sean Tobyne
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA
| | - Alvaro Pascual-Leone
- Linus Health, Inc., 280 Summer Street, 10th Floor, Boston, MA, 02210, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA.
| |
Collapse
|
21
|
Dai Y, Hsu YC, Fernandes BS, Zhang K, Li X, Enduru N, Liu A, Manuel AM, Jiang X, Zhao Z. Disentangling Accelerated Cognitive Decline from the Normal Aging Process and Unraveling Its Genetic Components: A Neuroimaging-Based Deep Learning Approach. J Alzheimers Dis 2024; 97:1807-1827. [PMID: 38306043 PMCID: PMC11649026 DOI: 10.3233/jad-231020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Background The progressive cognitive decline, an integral component of Alzheimer's disease (AD), unfolds in tandem with the natural aging process. Neuroimaging features have demonstrated the capacity to distinguish cognitive decline changes stemming from typical brain aging and AD between different chronological points. Objective To disentangle the normal aging effect from the AD-related accelerated cognitive decline and unravel its genetic components using a neuroimaging-based deep learning approach. Methods We developed a deep-learning framework based on a dual-loss Siamese ResNet network to extract fine-grained information from the longitudinal structural magnetic resonance imaging (MRI) data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study. We then conducted genome-wide association studies (GWAS) and post-GWAS analyses to reveal the genetic basis of AD-related accelerated cognitive decline. Results We used our model to process data from 1,313 individuals, training it on 414 cognitively normal people and predicting cognitive assessment for all participants. In our analysis of accelerated cognitive decline GWAS, we identified two genome-wide significant loci: APOE locus (chromosome 19 p13.32) and rs144614292 (chromosome 11 p15.1). Variant rs144614292 (G > T) has not been reported in previous AD GWA studies. It is within the intronic region of NELL1, which is expressed in neurons and plays a role in controlling cell growth and differentiation. The cell-type-specific enrichment analysis and functional enrichment of GWAS signals highlighted the microglia and immune-response pathways. Conclusions Our deep learning model effectively extracted relevant neuroimaging features and predicted individual cognitive decline. We reported a novel variant (rs144614292) within the NELL1 gene.
Collapse
Affiliation(s)
- Yulin Dai
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Yu-Chun Hsu
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Brisa S. Fernandes
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Kai Zhang
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Xiaoyang Li
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Biostatistics and Data Science, School of
Public Health, The University of Texas Health Science Center at Houston, Houston,
TX, USA
| | - Nitesh Enduru
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
| | - Andi Liu
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
| | - Astrid M. Manuel
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Xiaoqian Jiang
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
- Department of Biomedical Informatics, Vanderbilt University
Medical enter, Nashville, TN, USA
| |
Collapse
|
22
|
Nithya VP, Mohanasundaram N, Santhosh R. An Early Detection and Classification of Alzheimer's Disease Framework Based on ResNet-50. Curr Med Imaging 2024; 20:e250823220361. [PMID: 37622561 DOI: 10.2174/1573405620666230825113344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023]
Abstract
OBJECTIVE The objective of this study is to develop a more effective early detection system for Alzheimer's disease (AD) using a Deep Residual Network (ResNet) model by addressing the issue of convolutional layers in conventional Convolutional Neural Networks (CNN) and applying image preprocessing techniques. METHODS The proposed method involves using Contrast Limited Adaptive Histogram Equalizer (CLAHE) and Boosted Anisotropic Diffusion Filters (BADF) for equalization and noise removal and K-means clustering for segmentation. A ResNet-50 model with shortcut links between three residual layers is proposed to extract features more efficiently. ResNet-50 is preferred over other ResNet types due to its intermediate depth, striking a balance between computational efficiency and improved performance, making it a widely adopted and effective architecture for various computer vision tasks. While other ResNet variations may offer higher depths, they are more prone to overfitting and computational complexity, which can hinder their practical application. The proposed method is evaluated on a dataset of MRI scans of AD patients. RESULTS The proposed method achieved high accuracy and minimum losses of 95% and 0.12, respectively. While some models showed better accuracy, they were prone to overfitting. In contrast, the suggested framework, based on the ResNet-50 model, demonstrated superior performance in terms of various performance metrics, providing a robust and reliable approach to Alzheimer's disease categorization. CONCLUSION The proposed ResNet-50 model with shortcut links between three residual layers, combined with image preprocessing techniques, provides an effective early detection system for AD. The study demonstrates the potential of deep learning and image processing techniques in developing accurate and efficient diagnostic tools for AD. The proposed method improves the existing approaches to AD classification and provides a promising framework for future research in this area.
Collapse
Affiliation(s)
- V P Nithya
- Department of Computer Science and Engineering, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - N Mohanasundaram
- Department of Computer Science and Engineering, Faculty 0f Engineering, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - R Santhosh
- Department of Computer Science and Engineering, Faculty 0f Engineering, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| |
Collapse
|
23
|
Aghdam MA, Bozdag S, Saeed F. PVTAD: ALZHEIMER'S DISEASE DIAGNOSIS USING PYRAMID VISION TRANSFORMER APPLIED TO WHITE MATTER OF T1-WEIGHTED STRUCTURAL MRI DATA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567617. [PMID: 38045324 PMCID: PMC10690181 DOI: 10.1101/2023.11.17.567617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, and timely diagnosis is crucial for early interventions. AD is known to have disruptive local and global brain neural connections that may be instrumental in understanding and extracting specific biomarkers. Previous machine-learning approaches are mostly based on convolutional neural network (CNN) and standard vision transformer (ViT) models which may not sufficiently capture the multidimensional local and global patterns that may be indicative of AD. Therefore, in this paper, we propose a novel approach called PVTAD to classify AD and cognitively normal (CN) cases using pretrained pyramid vision transformer (PVT) and white matter (WM) of T1-weighted structural MRI (sMRI) data. Our approach combines the advantages of CNN and standard ViT to extract both local and global features indicative of AD from the WM coronal middle slices. We performed experiments on subjects with T1-weighed MPRAGE sMRI scans from the ADNI dataset. Our results demonstrate that the PVTAD achieves an average accuracy of 97.7% and F1-score of 97.6%, outperforming the single and parallel CNN and standard ViT architectures based on sMRI data for AD vs. CN classification.
Collapse
Affiliation(s)
- Maryam Akhavan Aghdam
- School of Computing and Information Sciences, Florida International University, Miami, FL, United States
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, United States
- Department of Mathematics, University of North Texas, Denton, TX, United States
- BioDiscovery Institute, University of North Texas, Denton, TX, United States
| | - Fahad Saeed
- School of Computing and Information Sciences, Florida International University, Miami, FL, United States
| |
Collapse
|
24
|
Gallagher RL, Koscik RL, Moody JF, Vogt NM, Adluru N, Kecskemeti SR, Van Hulle CA, Chin NA, Asthana S, Kollmorgen G, Suridjan I, Carlsson CM, Johnson SC, Dean DC, Zetterberg H, Blennow K, Alexander AL, Bendlin BB. Neuroimaging of tissue microstructure as a marker of neurodegeneration in the AT(N) framework: defining abnormal neurodegeneration and improving prediction of clinical status. Alzheimers Res Ther 2023; 15:180. [PMID: 37848950 PMCID: PMC10583332 DOI: 10.1186/s13195-023-01281-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 07/27/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Alzheimer's disease involves accumulating amyloid (A) and tau (T) pathology, and progressive neurodegeneration (N), leading to the development of the AD clinical syndrome. While several markers of N have been proposed, efforts to define normal vs. abnormal neurodegeneration based on neuroimaging have been limited. Sensitive markers that may account for or predict cognitive dysfunction for individuals in early disease stages are critical. METHODS Participants (n = 296) defined on A and T status and spanning the AD-clinical continuum underwent multi-shell diffusion-weighted magnetic resonance imaging to generate Neurite Orientation Dispersion and Density Imaging (NODDI) metrics, which were tested as markers of N. To better define N, we developed age- and sex-adjusted robust z-score values to quantify normal and AD-associated (abnormal) neurodegeneration in both cortical gray matter and subcortical white matter regions of interest. We used general logistic regression with receiver operating characteristic (ROC) and area under the curve (AUC) analysis to test whether NODDI metrics improved diagnostic accuracy compared to models that only relied on cerebrospinal fluid (CSF) A and T status (alone and in combination). RESULTS Using internal robust norms, we found that NODDI metrics correlate with worsening cognitive status and that NODDI captures early, AD neurodegenerative pathology in the gray matter of cognitively unimpaired, but A/T biomarker-positive, individuals. NODDI metrics utilized together with A and T status improved diagnostic prediction accuracy of AD clinical status, compared with models using CSF A and T status alone. CONCLUSION Using a robust norms approach, we show that abnormal AD-related neurodegeneration can be detected among cognitively unimpaired individuals. Metrics derived from diffusion-weighted imaging are potential sensitive markers of N and could be considered for trial enrichment and as outcomes in clinical trials. However, given the small sample sizes, the exploratory nature of the work must be acknowledged.
Collapse
Affiliation(s)
- Rigina L Gallagher
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
| | - Rebecca Langhough Koscik
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Jason F Moody
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Nicholas M Vogt
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
| | - Nagesh Adluru
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Waisman Research Center, Madison, WI, USA
| | | | - Carol A Van Hulle
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
| | - Nathaniel A Chin
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
| | - Sanjay Asthana
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Veterans Administration, Madison, WI, USA
| | | | | | - Cynthia M Carlsson
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Wisconsin Alzheimer's Institute, Madison, WI, USA
- Veterans Administration, Madison, WI, USA
| | - Sterling C Johnson
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Wisconsin Alzheimer's Institute, Madison, WI, USA
- Veterans Administration, Madison, WI, USA
| | - Douglas C Dean
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Waisman Research Center, Madison, WI, USA
| | - Henrik Zetterberg
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Andrew L Alexander
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Waisman Research Center, Madison, WI, USA
| | - Barbara B Bendlin
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA.
- Wisconsin Alzheimer's Institute, Madison, WI, USA.
| |
Collapse
|
25
|
Yang X, Qu H. Bibliometric review on biomarkers for Alzheimer's disease between 2000 and 2023. Medicine (Baltimore) 2023; 102:e34982. [PMID: 37682187 PMCID: PMC10489337 DOI: 10.1097/md.0000000000034982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a common cause of dementia and frailty. Therefore, it is important to develop biomarkers that can diagnose these changes to improve the likelihood of monitoring and treating potential causes. Therefore, this study aimed to examine the relationship between biomarkers and AD, identify journal publications and collaborators, and analyze keywords and research trends using a bibliometric method. METHODS We systematically searched for papers published in the Web of Science Core Collection database on biomarkers and AD. The search strategy was as follows: (TS) = (Alzheimer's OR Alzheimer's OR Alzheimer OR "Alzheimer's disease" OR "Alzheimer disease") AND TS = (biomarker OR biomarkers). Only articles and reviews were included as document types, with English as the primary language. The CiteSpace software was used to analyze the retrieved data on countries/regions, institutions, authors, published journals, and keywords. Simultaneously, the co-occurrence of the keywords was constructed. RESULTS There were 2625 articles on biomarkers and AD research published by 51 institutions located in 41 countries in 75 journals; the number of articles has shown an increasing trend over the past 20 years. Keywords analysis showed that Alzheimer's disease, cerebrospinal fluid, mild cognitive impairment, amyloid beta, and tau were also highly influential. CONCLUSION This was the first study to provide an overview of the current status of development, hot spots of study, and future trends in biomarkers for AD. These findings will provide useful information for researchers to explore trends and gaps in the field of biomarkers and AD.
Collapse
Affiliation(s)
- Xiaojie Yang
- Department of The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Huiling Qu
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
26
|
Dai Y, Yu-Chun H, Fernandes BS, Zhang K, Xiaoyang L, Enduru N, Liu A, Manuel AM, Jiang X, Zhao Z. Disentangling accelerated cognitive decline from the normal aging process and unraveling its genetic components: A neuroimaging-based deep learning approach. RESEARCH SQUARE 2023:rs.3.rs-3328861. [PMID: 37720047 PMCID: PMC10503860 DOI: 10.21203/rs.3.rs-3328861/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Background The progressive cognitive decline that is an integral component of AD unfolds in tandem with the natural aging process. Neuroimaging features have demonstrated the capacity to distinguish cognitive decline changes stemming from typical brain aging and Alzheimer's disease between different chronological points. Methods We developed a deep-learning framework based on dual-loss Siamese ResNet network to extract fine-grained information from the longitudinal structural magnetic resonance imaging (MRI) data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study. We then conducted genome-wide association studies (GWAS) and post-GWAS analyses to reveal the genetic basis of AD-related accelerated cognitive decline. Results We used our model to process data from 1,313 individuals, training it on 414 cognitively normal people and predicting cognitive assessment for all participants. In our analysis of accelerated cognitive decline GWAS, we identified two genome-wide significant loci: APOE locus (chromosome 19 p13.32) and rs144614292 (chromosome 11 p15.1). Variant rs144614292 (G>T) has not been reported in previous AD GWA studies. It is within the intronic region of NELL1, which is expressed in neuron and plays a role in controlling cell growth and differentiation. In addition, MUC7 and PROL1/OPRPNon chromosome 4 were significant at the gene level. The cell-type-specific enrichment analysis and functional enrichment of GWAS signals highlighted the microglia and immune-response pathways. Furthermore, we found that the cognitive decline slope GWAS was positively correlated with previous AD GWAS. Conclusion Our deep learning model was demonstrated effective on extracting relevant neuroimaging features and predicting individual cognitive decline. We reported a novel variant (rs144614292) within the NELL1 gene. Our approach has the potential to disentangle accelerated cognitive decline from the normal aging process and to determine its related genetic factors, leveraging opportunities for early intervention.
Collapse
Affiliation(s)
- Yulin Dai
- The University of Texas Health Science Center at Houston
| | - Hsu Yu-Chun
- The University of Texas Health Science Center at Houston
| | | | - Kai Zhang
- The University of Texas Health Science Center at Houston
| | - Li Xiaoyang
- The University of Texas Health Science Center at Houston
| | - Nitesh Enduru
- The University of Texas Health Science Center at Houston
| | - Andi Liu
- The University of Texas Health Science Center at Houston
| | | | - Xiaoqian Jiang
- The University of Texas Health Science Center at Houston
| | - Zhongming Zhao
- The University of Texas Health Science Center at Houston
| |
Collapse
|
27
|
Chatanaka MK, Sohaei D, Diamandis EP, Prassas I. Beyond the amyloid hypothesis: how current research implicates autoimmunity in Alzheimer's disease pathogenesis. Crit Rev Clin Lab Sci 2023; 60:398-426. [PMID: 36941789 DOI: 10.1080/10408363.2023.2187342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
The amyloid hypothesis has so far been at the forefront of explaining the pathogenesis of Alzheimer's Disease (AD), a progressive neurodegenerative disorder that leads to cognitive decline and eventual death. Recent evidence, however, points to additional factors that contribute to the pathogenesis of this disease. These include the neurovascular hypothesis, the mitochondrial cascade hypothesis, the inflammatory hypothesis, the prion hypothesis, the mutational accumulation hypothesis, and the autoimmunity hypothesis. The purpose of this review was to briefly discuss the factors that are associated with autoimmunity in humans, including sex, the gut and lung microbiomes, age, genetics, and environmental factors. Subsequently, it was to examine the rise of autoimmune phenomena in AD, which can be instigated by a blood-brain barrier breakdown, pathogen infections, and dysfunction of the glymphatic system. Lastly, it was to discuss the various ways by which immune system dysregulation leads to AD, immunomodulating therapies, and future directions in the field of autoimmunity and neurodegeneration. A comprehensive account of the recent research done in the field was extracted from PubMed on 31 January 2022, with the keywords "Alzheimer's disease" and "autoantibodies" for the first search input, and "Alzheimer's disease" with "IgG" for the second. From the first search, 19 papers were selected, because they contained recent research on the autoantibodies found in the biofluids of patients with AD. From the second search, four papers were selected. The analysis of the literature has led to support the autoimmune hypothesis in AD. Autoantibodies were found in biofluids (serum/plasma, cerebrospinal fluid) of patients with AD with multiple methods, including ELISA, Mass Spectrometry, and microarray analysis. Through continuous research, the understanding of the synergistic effects of the various components that lead to AD will pave the way for better therapeutic methods and a deeper understanding of the disease.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Dorsa Sohaei
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| |
Collapse
|
28
|
Pan F, Huang Y, Cai X, Wang Y, Guan Y, Deng J, Yang D, Zhu J, Zhao Y, Xie F, Fang Z, Guo Q. Integrated algorithm combining plasma biomarkers and cognitive assessments accurately predicts brain β-amyloid pathology. COMMUNICATIONS MEDICINE 2023; 3:65. [PMID: 37165172 PMCID: PMC10172320 DOI: 10.1038/s43856-023-00295-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Accurate prediction of cerebral amyloidosis with easily available indicators is urgently needed for diagnosis and treatment of Alzheimer's disease (AD). METHODS We examined plasma Aβ42, Aβ40, T-tau, P-tau181, and NfL, with APOE genotypes, cognitive test scores and key demographics in a large Chinese cohort (N = 609, aged 40 to 84 years) covering full AD spectrum. Data-driven integrated computational models were developed to predict brain β-amyloid (Aβ) pathology. RESULTS Our computational models accurately predict brain Aβ positivity (area under the ROC curves (AUC) = 0.94). The results are validated in Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Particularly, the models have the highest prediction power (AUC = 0.97) in mild cognitive impairment (MCI) participants. Three levels of models are designed with different accuracies and complexities. The model which only consists of plasma biomarkers can predict Aβ positivity in amnestic MCI (aMCI) patients with AUC = 0.89. Generally the models perform better in participants without comorbidities or family histories. CONCLUSIONS The innovative integrated models provide opportunity to assess Aβ pathology in a non-invasive and cost-effective way, which might facilitate AD-drug development, early screening, clinical diagnosis and prognosis evaluation.
Collapse
Affiliation(s)
- Fengfeng Pan
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yanlu Huang
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiao Cai
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Ying Wang
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiale Deng
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Dake Yang
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Jinhang Zhu
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Yike Zhao
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Zhuo Fang
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China.
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
29
|
Aramadaka S, Mannam R, Sankara Narayanan R, Bansal A, Yanamaladoddi VR, Sarvepalli SS, Vemula SL. Neuroimaging in Alzheimer's Disease for Early Diagnosis: A Comprehensive Review. Cureus 2023; 15:e38544. [PMID: 37273363 PMCID: PMC10239271 DOI: 10.7759/cureus.38544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, affecting roughly half of those over the age of 85. We briefly discussed the risk factors, epidemiology, and treatment options for AD. The development of therapeutic therapies operating very early in the disease cascade has been spurred by the realization that the disease process begins at least a decade or more before the manifestation of symptoms. Thus, the clinical significance of early diagnosis was emphasized. Using various keywords, a literature search was carried out using PubMed and other databases. For inclusion, pertinent articles were chosen and reviewed. This article has reviewed different neuroimaging techniques that are considered advanced tools to aid in establishing a diagnosis and highlighted the advantages as well as disadvantages of those techniques. Besides, the prevalence of several in vivo biomarkers aided in discriminating affected individuals from healthy controls in the early stages of the disease. Each imaging method has its advantages and disadvantages, hence no single imaging approach can be the optimum modality for diagnosis. This article also commented on a better approach to using these techniques to increase the likelihood of an early diagnosis.
Collapse
Affiliation(s)
| | - Raam Mannam
- Research, Narayana Medical College, Nellore, IND
| | | | - Arpit Bansal
- Research, Narayana Medical College, Nellore, IND
| | | | | | | |
Collapse
|
30
|
Mirkin S, Albensi BC. Should artificial intelligence be used in conjunction with Neuroimaging in the diagnosis of Alzheimer's disease? Front Aging Neurosci 2023; 15:1094233. [PMID: 37187577 PMCID: PMC10177660 DOI: 10.3389/fnagi.2023.1094233] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/27/2023] [Indexed: 05/17/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disorder that affects memory, thinking, behavior, and other cognitive functions. Although there is no cure, detecting AD early is important for the development of a therapeutic plan and a care plan that may preserve cognitive function and prevent irreversible damage. Neuroimaging, such as magnetic resonance imaging (MRI), computed tomography (CT), and positron emission tomography (PET), has served as a critical tool in establishing diagnostic indicators of AD during the preclinical stage. However, as neuroimaging technology quickly advances, there is a challenge in analyzing and interpreting vast amounts of brain imaging data. Given these limitations, there is great interest in using artificial Intelligence (AI) to assist in this process. AI introduces limitless possibilities in the future diagnosis of AD, yet there is still resistance from the healthcare community to incorporate AI in the clinical setting. The goal of this review is to answer the question of whether AI should be used in conjunction with neuroimaging in the diagnosis of AD. To answer the question, the possible benefits and disadvantages of AI are discussed. The main advantages of AI are its potential to improve diagnostic accuracy, improve the efficiency in analyzing radiographic data, reduce physician burnout, and advance precision medicine. The disadvantages include generalization and data shortage, lack of in vivo gold standard, skepticism in the medical community, potential for physician bias, and concerns over patient information, privacy, and safety. Although the challenges present fundamental concerns and must be addressed when the time comes, it would be unethical not to use AI if it can improve patient health and outcome.
Collapse
Affiliation(s)
- Sophia Mirkin
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Benedict C. Albensi
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
- St. Boniface Hospital Research, Winnipeg, MB, Canada
- University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
31
|
Swinford CG, Risacher SL, Wu YC, Apostolova LG, Gao S, Bice PJ, Saykin AJ. Altered cerebral blood flow in older adults with Alzheimer's disease: a systematic review. Brain Imaging Behav 2023; 17:223-256. [PMID: 36484922 PMCID: PMC10117447 DOI: 10.1007/s11682-022-00750-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/26/2022] [Accepted: 11/20/2022] [Indexed: 12/13/2022]
Abstract
The prevalence of Alzheimer's disease is projected to reach 13 million in the U.S. by 2050. Although major efforts have been made to avoid this outcome, so far there are no treatments that can stop or reverse the progressive cognitive decline that defines Alzheimer's disease. The utilization of preventative treatment before significant cognitive decline has occurred may ultimately be the solution, necessitating a reliable biomarker of preclinical/prodromal disease stages to determine which older adults are most at risk. Quantitative cerebral blood flow is a promising potential early biomarker for Alzheimer's disease, but the spatiotemporal patterns of altered cerebral blood flow in Alzheimer's disease are not fully understood. The current systematic review compiles the findings of 81 original studies that compared resting gray matter cerebral blood flow in older adults with mild cognitive impairment or Alzheimer's disease and that of cognitively normal older adults and/or assessed the relationship between cerebral blood flow and objective cognitive function. Individuals with Alzheimer's disease had relatively decreased cerebral blood flow in all brain regions investigated, especially the temporoparietal and posterior cingulate, while individuals with mild cognitive impairment had consistent results of decreased cerebral blood flow in the posterior cingulate but more mixed results in other regions, especially the frontal lobe. Most papers reported a positive correlation between regional cerebral blood flow and cognitive function. This review highlights the need for more studies assessing cerebral blood flow changes both spatially and temporally over the course of Alzheimer's disease, as well as the importance of including potential confounding factors in these analyses.
Collapse
Affiliation(s)
- Cecily G Swinford
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liana G Apostolova
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paula J Bice
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
32
|
Poppell M, Hammel G, Ren Y. Immune Regulatory Functions of Macrophages and Microglia in Central Nervous System Diseases. Int J Mol Sci 2023; 24:5925. [PMID: 36982999 PMCID: PMC10059890 DOI: 10.3390/ijms24065925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Macrophages can be characterized as a very multifunctional cell type with a spectrum of phenotypes and functions being observed spatially and temporally in various disease states. Ample studies have now demonstrated a possible causal link between macrophage activation and the development of autoimmune disorders. How these cells may be contributing to the adaptive immune response and potentially perpetuating the progression of neurodegenerative diseases and neural injuries is not fully understood. Within this review, we hope to illustrate the role that macrophages and microglia play as initiators of adaptive immune response in various CNS diseases by offering evidence of: (1) the types of immune responses and the processes of antigen presentation in each disease, (2) receptors involved in macrophage/microglial phagocytosis of disease-related cell debris or molecules, and, finally, (3) the implications of macrophages/microglia on the pathogenesis of the diseases.
Collapse
Affiliation(s)
| | | | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
33
|
Tio ES, Hohman TJ, Milic M, Bennett DA, Felsky D. Testing a polygenic risk score for morphological microglial activation in Alzheimer's disease and aging. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.10.23287119. [PMID: 36993775 PMCID: PMC10055438 DOI: 10.1101/2023.03.10.23287119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Neuroinflammation and the activation of microglial cells are among the earliest events in Alzheimer's disease (AD). However, direct observation of microglia in living people is not currently possible. Here, we indexed the heritable propensity for neuroinflammation with polygenic risk scores (PRS), using results from a recent genome-wide analysis of a validated post-mortem measure of morphological microglial activation. We sought to determine whether a PRS for microglial activation (PRS mic ) could augment the predictive performance of existing AD PRSs for late-life cognitive impairment. First, PRS mic were calculated and optimized in a calibration cohort (Alzheimer's Disease Neuroimaging Initiative (ADNI), n=450), with resampling. Second, predictive performance of optimal PRS mic was assessed in two independent, population-based cohorts (total n=212,237). Our PRS mic showed no significant improvement in predictive power for either AD diagnosis or cognitive performance. Finally, we explored associations of PRS mic with a comprehensive set of imaging and fluid AD biomarkers in ADNI. This revealed some nominal associations, but with inconsistent effect directions. While genetic scores capable of indexing risk for neuroinflammatory processes in aging are highly desirable, more well-powered genome-wide studies of microglial activation are required. Further, biobank-scale studies would benefit from phenotyping of proximal neuroinflammatory processes to improve the PRS development phase.
Collapse
Affiliation(s)
- Earvin S. Tio
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, CANADA
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, CANADA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Centre, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Milos Milic
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, CANADA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
| | - Daniel Felsky
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, CANADA
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, CANADA
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, CANADA
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, CANADA
| |
Collapse
|
34
|
Advanced Overview of Biomarkers and Techniques for Early Diagnosis of Alzheimer's Disease. Cell Mol Neurobiol 2023:10.1007/s10571-023-01330-y. [PMID: 36847930 DOI: 10.1007/s10571-023-01330-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023]
Abstract
The development of early non-invasive diagnosis methods and identification of novel biomarkers are necessary for managing Alzheimer's disease (AD) and facilitating effective prognosis and treatment. AD has multi-factorial nature and involves complex molecular mechanism, which causes neuronal degeneration. The primary challenges in early AD detection include patient heterogeneity and lack of precise diagnosis at the preclinical stage. Several cerebrospinal fluid (CSF) and blood biomarkers have been proposed to show excellent diagnosis ability by identifying tau pathology and cerebral amyloid beta (Aβ) for AD. Intense research endeavors are being made to develop ultrasensitive detection techniques and find potent biomarkers for early AD diagnosis. To mitigate AD worldwide, understanding various CSF biomarkers, blood biomarkers, and techniques that can be used for early diagnosis is imperative. This review attempts to provide information regarding AD pathophysiology, genetic and non-genetic factors associated with AD, several potential blood and CSF biomarkers, like neurofilament light, neurogranin, Aβ, and tau, along with biomarkers under development for AD detection. Besides, numerous techniques, such as neuroimaging, spectroscopic techniques, biosensors, and neuroproteomics, which are being explored to aid early AD detection, have been discussed. The insights thus gained would help in finding potential biomarkers and suitable techniques for the accurate diagnosis of early AD before cognitive dysfunction.
Collapse
|
35
|
Nasab AS, Noorani F, Paeizi Z, Khani L, Banaei S, Sadeghi M, Shafeghat M, Shafie M, Mayeli M, Initiative (ADNI) TADN. A Comprehensive Investigation of the Potential Role of Lipoproteins and Metabolite Profile as Biomarkers of Alzheimer's Disease Compared to the Known CSF Biomarkers. Int J Alzheimers Dis 2023; 2023:3540020. [PMID: 36936136 PMCID: PMC10019964 DOI: 10.1155/2023/3540020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 12/22/2022] [Accepted: 01/30/2023] [Indexed: 03/14/2023] Open
Abstract
Introduction While cerebrospinal fluid (CSF) core biomarkers have been considered diagnostic biomarkers for a long time, special attention has been recently dedicated to lipoproteins and metabolites that could be potentially associated with Alzheimer's disease (AD) neurodegeneration. Herein, we aimed to investigate the relationship between the levels of CSF core biomarkers including Aβ-42, TAU, and P-TAU and plasma lipoproteins and metabolites of patients with AD from the baseline cohort of the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Method Using the ADNI database, fourteen subclasses of lipoproteins as well as a number of lipids and fatty acids and low-molecular metabolites including amino acids, ketone bodies, and glycolysis-related metabolites in blood samples were measured as potential noninvasive markers, and their association with the CSF core biomarkers was statistically investigated controlling for age and gender. Results A total number of 251 AD subjects were included, among whom 71 subjects were negative for the Apo-E ε4 allele and 150 were positive. There was no significant difference between the two groups regarding cognitive assessments, CSF core biomarkers, and lipoproteins and metabolites except the level of Aβ-42 (p < 0.001) and phenylalanine (p = 0.049), which were higher in the negative group. CSF TAU and P-TAU were significantly correlated with medium and small HDL in the negative group, and with extremely large VLDL in the positive group. Our results also indicated significant correlations of metabolites including unsaturated fatty acids, glycerol, and leucine with CSF core biomarkers. Conclusion Based on our findings, a number of lipoproteins and metabolites were associated with CSF core biomarkers of AD. These correlations showed some differences in Apo-E ε4 positive and negative groups, which reminds the role of Apo-E gene status in the pathophysiology of AD development. However, further research is warranted to explore the exact association of lipoproteins and other metabolites with AD core biomarkers and pathology.
Collapse
Affiliation(s)
- Azam Sajjadi Nasab
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Noorani
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Paeizi
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Khani
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saba Banaei
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadeghi
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Melika Shafeghat
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- 2School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahan Shafie
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- 2School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Mayeli
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- 2School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
36
|
Löscher W, Stafstrom CE. Epilepsy and its neurobehavioral comorbidities: Insights gained from animal models. Epilepsia 2023; 64:54-91. [PMID: 36197310 DOI: 10.1111/epi.17433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023]
Abstract
It is well established that epilepsy is associated with numerous neurobehavioral comorbidities, with a bidirectional relationship; people with epilepsy have an increased incidence of depression, anxiety, learning and memory difficulties, and numerous other psychosocial challenges, and the occurrence of epilepsy is higher in individuals with those comorbidities. Although the cause-and-effect relationship is uncertain, a fuller understanding of the mechanisms of comorbidities within the epilepsies could lead to improved therapeutics. Here, we review recent data on epilepsy and its neurobehavioral comorbidities, discussing mainly rodent models, which have been studied most extensively, and emphasize that clinically relevant information can be gained from preclinical models. Furthermore, we explore the numerous potential factors that may confound the interpretation of emerging data from animal models, such as the specific seizure induction method (e.g., chemical, electrical, traumatic, genetic), the role of species and strain, environmental factors (e.g., laboratory environment, handling, epigenetics), and the behavioral assays that are chosen to evaluate the various aspects of neural behavior and cognition. Overall, the interplay between epilepsy and its neurobehavioral comorbidities is undoubtedly multifactorial, involving brain structural changes, network-level differences, molecular signaling abnormalities, and other factors. Animal models are well poised to help dissect the shared pathophysiological mechanisms, neurological sequelae, and biomarkers of epilepsy and its comorbidities.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
37
|
Yuan W, Zhi W, Ma L, Hu X, Wang Q, Zou Y, Wang L. Neural Oscillation Disorder in the Hippocampal CA1 Region of Different Alzheimer's Disease Mice. Curr Alzheimer Res 2023; 20:350-359. [PMID: 37559542 PMCID: PMC10661967 DOI: 10.2174/1567205020666230808122643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/16/2023] [Accepted: 06/30/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a well-known neurodegenerative disease that gradually induces neural network dysfunction and progressive memory deficits. Neural network activity is represented by rhythmic oscillations that influence local field potentials (LFPs). However, changes in hippocampal neural rhythmic oscillations in the early stage of AD remain largely unexplored. OBJECTIVE This study investigated neural rhythmic oscillations in 3-month-old APP/PS1 and 5x- FAD mice to assess early neural connectivity in AD. METHODS LFPs were recorded from the hippocampal CA1 region with implanted microelectrode arrays while the mice were in the awake resting stage. Welch fast Fourier transforms, continuous wavelet transforms, and phase-amplitude coupling analyses were used to compute the power density of different frequency bands and phase-amplitude modulation indices in the LFPs. RESULTS Our results showed impaired theta, low gamma, and high gamma frequency band power in APP/PS1 and 5xFAD mice during the awake resting stage. AD mice also showed decreased delta, alpha, and beta frequency band power. Impaired theta-low gamma and theta-high gamma phaseamplitude coupling were observed in 5xFAD mice. CONCLUSION This study revealed neural network activity differences in oscillation power and cross-frequency coupling in the early stage of AD, providing a new perspective for developing biomarkers for early AD diagnosis.
Collapse
Affiliation(s)
- Weiming Yuan
- Graduate Collaborative Training Base of Academy of Military Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Weijia Zhi
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Lizhen Ma
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Xiangjun Hu
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Qian Wang
- Department of Medical Imaging, Chinese PAP Beijing Corps Hospital, Beijing 100600, China
| | - Yong Zou
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Lifeng Wang
- Graduate Collaborative Training Base of Academy of Military Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| |
Collapse
|
38
|
Tio ES, Hohman TJ, Milic M, Bennett DA, Felsky D. Testing a Polygenic Risk Score for Morphological Microglial Activation in Alzheimer's Disease and Aging. J Alzheimers Dis 2023; 94:1549-1561. [PMID: 37458040 PMCID: PMC11062501 DOI: 10.3233/jad-230434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
BACKGROUND Neuroinflammation and the activation of microglial cells are among the earliest events in Alzheimer's disease (AD). However, direct observation of microglia in living people is not currently possible. Here, we indexed the heritable propensity for neuroinflammation with polygenic risk scores (PRS), using results from a recent genome-wide analysis of a validated post-mortem measure of morphological microglial activation. OBJECTIVE We sought to determine whether a PRS for microglial activation (PRSmic) could augment the predictive performance of existing AD PRSs for late-life cognitive impairment. METHODS First, PRSmic were calculated and optimized in a calibration cohort (Alzheimer's Disease Neuroimaging Initiative (ADNI), n = 450), with resampling. Second, predictive performance of optimal PRSmic was assessed in two independent, population-based cohorts (total n = 212,237). Finally, we explored associations of PRSmic with a comprehensive set of imaging and fluid AD biomarkers in ADNI. RESULTS Our PRSmic showed no significant improvement in predictive power for either AD diagnosis or cognitive performance in either external cohort. Some nominal associations were found in ADNI, but with inconsistent effect directions. CONCLUSION While genetic scores capable of indexing risk for neuroinflammatory processes in aging are highly desirable, more well-powered genome-wide studies of microglial activation are required. Further, biobank-scale studies would benefit from phenotyping of proximal neuroinflammatory processes to improve the PRS development phase.
Collapse
Affiliation(s)
- Earvin S. Tio
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, CANADA
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, CANADA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Centre, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Milos Milic
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, CANADA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
| | - Daniel Felsky
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, CANADA
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, CANADA
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, CANADA
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, CANADA
| | | |
Collapse
|
39
|
Youn YC, Kim HR, Shin HW, Jeong HB, Han SW, Pyun JM, Ryoo N, Park YH, Kim S. Prediction of amyloid PET positivity via machine learning algorithms trained with EDTA-based blood amyloid-β oligomerization data. BMC Med Inform Decis Mak 2022; 22:286. [DOI: 10.1186/s12911-022-02024-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
The tendency of amyloid-β to form oligomers in the blood as measured with Multimer Detection System-Oligomeric Amyloid-β (MDS-OAβ) is a valuable biomarker for Alzheimer’s disease and has been verified with heparin-based plasma. The objective of this study was to evaluate the performance of ethylenediaminetetraacetic acid (EDTA)-based MDS-OAβ and to develop machine learning algorithms to predict amyloid positron emission tomography (PET) positivity.
Methods
The performance of EDTA-based MDS-OAβ in predicting PET positivity was evaluated in 312 individuals with various machine learning models. The models with various combinations of features (i.e., MDS-OAβ level, age, apolipoprotein E4 alleles, and Mini-Mental Status Examination [MMSE] score) were tested 50 times on each dataset.
Results
The random forest model best-predicted amyloid PET positivity based on MDS-OAβ combined with other features with an accuracy of 77.14 ± 4.21% and an F1 of 85.44 ± 3.10%. The order of significance of predictive features was MDS-OAβ, MMSE, Age, and APOE. The Support Vector Machine using the MDS-OAβ value only showed an accuracy of 71.09 ± 3.27% and F−1 value of 80.18 ± 2.70%.
Conclusions
The Random Forest model using EDTA-based MDS-OAβ combined with the MMSE and apolipoprotein E status can be used to prescreen for amyloid PET positivity.
Collapse
|
40
|
Sone D, Beheshti I. Neuroimaging-Based Brain Age Estimation: A Promising Personalized Biomarker in Neuropsychiatry. J Pers Med 2022; 12:jpm12111850. [PMID: 36579560 PMCID: PMC9695293 DOI: 10.3390/jpm12111850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 11/10/2022] Open
Abstract
It is now possible to estimate an individual's brain age via brain scans and machine-learning models. This validated technique has opened up new avenues for addressing clinical questions in neurology, and, in this review, we summarize the many clinical applications of brain-age estimation in neuropsychiatry and general populations. We first provide an introduction to typical neuroimaging modalities, feature extraction methods, and machine-learning models that have been used to develop a brain-age estimation framework. We then focus on the significant findings of the brain-age estimation technique in the field of neuropsychiatry as well as the usefulness of the technique for addressing clinical questions in neuropsychiatry. These applications may contribute to more timely and targeted neuropsychiatric therapies. Last, we discuss the practical problems and challenges described in the literature and suggest some future research directions.
Collapse
Affiliation(s)
- Daichi Sone
- Department of Psychiatry, Jikei University School of Medicine, Tokyo 105-8461, Japan
- Correspondence: ; Tel.: +81-03-3433
| | - Iman Beheshti
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
| |
Collapse
|
41
|
Aplicaciones de aprendizaje automático en salud. REVISTA MÉDICA CLÍNICA LAS CONDES 2022. [DOI: 10.1016/j.rmclc.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
42
|
Saunders TS, Gadd DA, Spires‐Jones TL, King D, Ritchie C, Muniz‐Terrera G. Associations between cerebrospinal fluid markers and cognition in ageing and dementia: A systematic review. Eur J Neurosci 2022; 56:5650-5713. [PMID: 35338546 PMCID: PMC9790745 DOI: 10.1111/ejn.15656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 12/30/2022]
Abstract
A biomarker associated with cognition in neurodegenerative dementias would aid in the early detection of disease progression, complement clinical staging and act as a surrogate endpoint in clinical trials. The current systematic review evaluates the association between cerebrospinal fluid protein markers of synapse loss and neuronal injury and cognition. We performed a systematic search which revealed 67 studies reporting an association between cerebrospinal fluid markers of interest and neuropsychological performance. Despite the substantial heterogeneity between studies, we found some evidence for an association between neurofilament-light and worse cognition in Alzheimer's diseases, frontotemporal dementia and typical cognitive ageing. Moreover, there was an association between cerebrospinal fluid neurogranin and cognition in those with an Alzheimer's-like cerebrospinal fluid biomarker profile. Some evidence was found for cerebrospinal fluid neuronal pentraxin-2 as a correlate of cognition across dementia syndromes. Due to the substantial heterogeneity of the field, no firm conclusions can be drawn from this review. Future research should focus on improving standardization and reporting as well as establishing the importance of novel markers such as neuronal pentraxin-2 and whether such markers can predict longitudinal cognitive decline.
Collapse
Affiliation(s)
- Tyler S. Saunders
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| | - Danni A. Gadd
- Center for Genomic and Experimental Medicine, Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Tara L. Spires‐Jones
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - Declan King
- UK Dementia Research InstituteThe University of EdinburghEdinburghUK
- Center for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - Craig Ritchie
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| | - Graciela Muniz‐Terrera
- Center for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Center for Dementia PreventionThe University of EdinburghEdinburghUK
| |
Collapse
|
43
|
Bunnik EM, Smedinga M, Milne R, Georges J, Richard E, Schermer MHN. Ethical Frameworks for Disclosure of Alzheimer Disease Biomarkers to Research Participants: Conflicting Norms and a Nuanced Policy. Ethics Hum Res 2022; 44:2-13. [PMID: 36316970 DOI: 10.1002/eahr.500146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
More and more frequently, clinical trials for Alzheimer disease (AD) are targeting cognitively unimpaired individuals who are at increased risk of developing the disease. It is not always clear whether AD biomarker information should be disclosed to research participants: on the one hand, research participants may be interested in learning this information because of its perceived utility, but on the other hand, learning this information may be harmful, as there are very few effective preventive or therapeutic options available for AD. In this article, we bring together three separate sets of ethical guidance literature: on the return of individual research results, on an individual's right to access personal data, and on transparent enrollment into clinical trials. Based on these literatures, we suggest policies for the disclosure of AD biomarker test results in longitudinal observational cohort studies, clinical trials, and hybrid research projects, such as the European Prevention of Alzheimer's Dementia (EPAD) project, in which we served as an ethics team. We also present and critically discuss recommendations for disclosure of AD biomarkers in practice. We underscore that, as long as the clinical validity of AD biomarkers remains limited, there are good reasons to avoid actively disclosing them to cognitively unimpaired research participants.
Collapse
Affiliation(s)
- Eline M Bunnik
- Associate professor at the Erasmus University Medical Center in the Department of Medical Ethics, Philosophy and History of Medicine
| | - Marthe Smedinga
- Scientific secretary for the subcommittee on ethics and societal aspects at the Netherlands Commission on Genetic Modification
| | - Richard Milne
- Sociologist of science, technology, and medicine, the head of research and dialogue at Wellcome Connecting Science, and the deputy director of the Kavli Centre for Ethics, Science, and the Public at the University of Cambridge
| | | | - Edo Richard
- Professor of neurology at Amsterdam University Medical Centre and a neurologist at the Department of Neurology at the Donders Institute for Brain, Cognition and Behaviour at Radboud University Medical Centre
| | - Maartje H N Schermer
- Professor of philosophy of medicine at the Erasmus University Medical Center in the Department of Ethics, Philosophy and History of Medicine
| |
Collapse
|
44
|
Schilling LP, Balthazar MLF, Radanovic M, Forlenza OV, Silagi ML, Smid J, Barbosa BJAP, Frota NAF, Souza LCD, Vale FAC, Caramelli P, Bertolucci PHF, Chaves MLF, Brucki SMD, Damasceno BP, Nitrini R. Diagnosis of Alzheimer’s disease: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022. [DOI: 10.1590/1980-5764-dn-2022-s102en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
ABSTRACT This paper presents the consensus of the Scientific Department of Cognitive Neurology and Aging from the Brazilian Academy of Neurology on the diagnostic criteria for Alzheimer’s disease (AD) in Brazil. The authors conducted a literature review regarding clinical and research criteria for AD diagnosis and proposed protocols for use at primary, secondary, and tertiary care levels. Within this clinical scenario, the diagnostic criteria for typical and atypical AD are presented as well as clinical, cognitive, and functional assessment tools and complementary propaedeutics with laboratory and neuroimaging tests. The use of biomarkers is also discussed for both clinical diagnosis (in specific conditions) and research.
Collapse
Affiliation(s)
- Lucas Porcello Schilling
- Pontifícia Universidade do Rio Grande do Sul, Brasil; Pontifícia Universidade do Rio Grande do Sul, Brasil; Pontifícia Universidade do Rio Grande do Sul, Brasil
| | | | | | | | - Marcela Lima Silagi
- Universidade Federal de São Paulo, Brasil; Universidade de São Paulo, Brasil
| | | | - Breno José Alencar Pires Barbosa
- Universidade de São Paulo, Brasil; Universidade Federal de Pernambuco, Brasil; Instituto de Medicina Integral Prof. Fernando Figueira, Brasil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Schilling LP, Balthazar MLF, Radanovic M, Forlenza OV, Silagi ML, Smid J, Barbosa BJAP, Frota NAF, de Souza LC, Vale FAC, Caramelli P, Bertolucci PHF, Chaves MLF, Brucki SMD, Damasceno BP, Nitrini R. Diagnosis of Alzheimer's disease: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022; 16:25-39. [PMID: 36533157 PMCID: PMC9745995 DOI: 10.1590/1980-5764-dn-2022-s102pt] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/22/2021] [Accepted: 04/27/2022] [Indexed: 01/25/2023] Open
Abstract
This paper presents the consensus of the Scientific Department of Cognitive Neurology and Aging from the Brazilian Academy of Neurology on the diagnostic criteria for Alzheimer's disease (AD) in Brazil. The authors conducted a literature review regarding clinical and research criteria for AD diagnosis and proposed protocols for use at primary, secondary, and tertiary care levels. Within this clinical scenario, the diagnostic criteria for typical and atypical AD are presented as well as clinical, cognitive, and functional assessment tools and complementary propaedeutics with laboratory and neuroimaging tests. The use of biomarkers is also discussed for both clinical diagnosis (in specific conditions) and research.
Collapse
Affiliation(s)
- Lucas Porcello Schilling
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brasil
- Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brasil
- Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brasil
| | | | - Márcia Radanovic
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brasil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brasil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Psiquiatria, São Paulo SP, Brasil
| | - Marcela Lima Silagi
- Universidade Federal de São Paulo, Departamento de Fonoaudiologia, São Paulo SP, Brasil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Jerusa Smid
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Breno José Alencar Pires Barbosa
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
- Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brasil
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brasil
| | | | - Leonardo Cruz de Souza
- Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brasil
| | - Francisco Assis Carvalho Vale
- Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brasil
| | - Paulo Caramelli
- Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brasil
| | | | - Márcia Lorena Fagundes Chaves
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brasil
| | - Sonia Maria Dozzi Brucki
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Benito Pereira Damasceno
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brasil
| | - Ricardo Nitrini
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| |
Collapse
|
46
|
Dulewicz M, Kulczyńska-Przybik A, Mroczko P, Kornhuber J, Lewczuk P, Mroczko B. Biomarkers for the Diagnosis of Alzheimer’s Disease in Clinical Practice: The Role of CSF Biomarkers during the Evolution of Diagnostic Criteria. Int J Mol Sci 2022; 23:ijms23158598. [PMID: 35955728 PMCID: PMC9369334 DOI: 10.3390/ijms23158598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/30/2022] [Accepted: 07/30/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive condition and the most common cause of dementia worldwide. The neuropathological changes characteristic of the disorder can be successfully detected before the development of full-blown AD. Early diagnosis of the disease constitutes a formidable challenge for clinicians. CSF biomarkers are the in vivo evidence of neuropathological changes developing in the brain of dementia patients. Therefore, measurement of their concentrations allows for improved accuracy of clinical diagnosis. Moreover, AD biomarkers may provide an indication of disease stage. Importantly, the CSF biomarkers of AD play a pivotal role in the new diagnostic criteria for the disease, and in the recent biological definition of AD by the National Institute on Aging, NIH and Alzheimer’s Association. Due to the necessity of collecting CSF by lumbar puncture, the procedure seems to be an important issue not only from a medical, but also a legal, viewpoint. Furthermore, recent technological advances may contribute to the automation of AD biomarkers measurement and may result in the establishment of unified cut-off values and reference limits. Moreover, a group of international experts in the field of AD biomarkers have developed a consensus and guidelines on the interpretation of CSF biomarkers in the context of AD diagnosis. Thus, technological advancement and expert recommendations may contribute to a more widespread use of these diagnostic tests in clinical practice to support a diagnosis of mild cognitive impairment (MCI) or dementia due to AD. This review article presents up-to-date data regarding the usefulness of CSF biomarkers in routine clinical practice and in biomarkers research.
Collapse
Affiliation(s)
- Maciej Dulewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (P.L.); (B.M.)
- Correspondence:
| | - Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (P.L.); (B.M.)
| | - Piotr Mroczko
- Department of Criminal Law and Criminology, Faculty of Law, University of Bialystok, 15-213 Bialystok, Poland;
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Piotr Lewczuk
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (P.L.); (B.M.)
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (P.L.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
47
|
Zhu LY, Shi L, Luo Y, Leung J, Kwok T. Brain MRI Biomarkers to Predict Cognitive Decline in Older People with Alzheimer's Disease. J Alzheimers Dis 2022; 88:763-769. [PMID: 35723095 DOI: 10.3233/jad-215189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Structural magnetic resonance imaging markers predicting symptomatic progression at the individual level can be highly beneficial for early intervention and treatment planning for Alzheimer's disease (AD). However, the correlation between baseline MRI findings and AD progression has not been fully established. OBJECTIVE To explore the correlation between baseline MRI findings and AD progression. METHODS Brain volumetric measures were applied to differentiate the patients at risk of fast deterioration in AD. We included 194 AD patients with a 24-month follow-up: 65 slow decliners, 63 normal decliners, and 66 fast decliners categorized by changes in Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog). ANOVA analyses were used to identify baseline brain atrophy between groups. Logistic regressions were further performed to explore the relative merits of AD resemblance structural atrophy index (AD-RAI) and individual regional volumetric measures in prediction of disease progression. RESULTS Atrophy in the temporal and insular lobes was associated with fast cognitive decline over 24 months. Smaller volumes of temporal and insular lobes in the left but not the right brain were associated with fast cognitive decline. Baseline AD-RAI predicted fast versus slow progression of cognitive decline (odds ratio 3.025 (95% CI: 1.064-8.600), high versus low, AUC 0.771). Moreover, AD-RAI was significantly lower among slow decliners when compared with normal decliners (p = 0.039). CONCLUSION AD-RAI on MRI showed potential in identifying clinical AD patients at risk of accelerated cognitive decline.
Collapse
Affiliation(s)
- Liu-Ying Zhu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Zhongshan City People's Hospital, Zhongshan, Guangdong Province, China
| | - Lin Shi
- BrainNow Research Institute, Shenzhen, Guangdong Province, China.,Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, China
| | - Yishan Luo
- BrainNow Research Institute, Shenzhen, Guangdong Province, China
| | - Jason Leung
- Jockey Club Centre for Osteoporosis Care and Control, The Chinese University of Hong Kong, Hong Kong, Shatin, China
| | - Timothy Kwok
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| |
Collapse
|
48
|
Abstract
Alzheimer’s Disease (AD) is a neurodegenerative disorder that is characterized clinically by progressive cognitive decline and pathologically by the β-sheet rich fibril plaque deposition of the amyloid-β (Aβ) peptide in the brain. While plaques are a hallmark of AD, plaque burden is not correlated with cognitive impairment. Instead, Aβ oligomers formed during the aggregation process represent the main agents of neurotoxicity, which occurs 10–20 years before patients begin to show symptoms. These oligomers are dynamic in nature and represented by a heterogeneous distribution of aggregates ranging from low- to high-molecular weight, some of which are toxic while others are not. A major difficulty in determining the pathological mechanism(s) of Aβ, developing reliable diagnostic markers for early-stage detection, as well as effective therapeutics for AD are the differentiation and characterization of oligomers formed throughout disease propagation based on their molecular features, effects on biological function, and relevance to disease propagation and pathology. Thus, it is critical to methodically identify the mechanisms of Aβ aggregation and toxicity, as well as describe the roles of different oligomers and aggregates in disease progression and molecular pathology. Here, we describe a variety of biophysical techniques used to isolate and characterize a range of Aβ oligomer populations, as well as discuss proposed mechanisms of toxicity and therapeutic interventions aimed at specific assemblies formed during the aggregation process. The approaches being used to map the misfolding and aggregation of Aβ are like what was done during the fundamental early studies, mapping protein folding pathways using combinations of biophysical techniques in concert with protein engineering. Such information is critical to the design and molecular engineering of future diagnostics and therapeutics for AD.
Collapse
|
49
|
Corbin D, Lesage F. Assessment of the predictive potential of cognitive scores from retinal images and retinal fundus metadata via deep learning using the CLSA database. Sci Rep 2022; 12:5767. [PMID: 35388080 PMCID: PMC8986784 DOI: 10.1038/s41598-022-09719-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 03/25/2022] [Indexed: 01/19/2023] Open
Abstract
Accumulation of beta-amyloid in the brain and cognitive decline are considered hallmarks of Alzheimer’s disease. Knowing from previous studies that these two factors can manifest in the retina, the aim was to investigate whether a deep learning method was able to predict the cognition of an individual from a RGB image of his retina and metadata. A deep learning model, EfficientNet, was used to predict cognitive scores from the Canadian Longitudinal Study on Aging (CLSA) database. The proposed model explained 22.4% of the variance in cognitive scores on the test dataset using fundus images and metadata. Metadata alone proved to be more effective in explaining the variance in the sample (20.4%) versus fundus images (9.3%) alone. Attention maps highlighted the optic nerve head as the most influential feature in predicting cognitive scores. The results demonstrate that RGB fundus images are limited in predicting cognition.
Collapse
Affiliation(s)
- Denis Corbin
- Laboratoire d'Imagerie optique et Moléculaire, Polytechnique Montréal, 2500 Chemin de Polytechnique Montréal, Montreal, QC, H3T 1J4, Canada.
| | - Frédéric Lesage
- Laboratoire d'Imagerie optique et Moléculaire, Polytechnique Montréal, 2500 Chemin de Polytechnique Montréal, Montreal, QC, H3T 1J4, Canada.,Institut de Cardiologie de Montréal, 5000 Rue Bélanger, Montreal, QC, H1T 1C8, Canada
| |
Collapse
|
50
|
Wei X, Du X, Xie Y, Suo X, He X, Ding H, Zhang Y, Ji Y, Chai C, Liang M, Yu C, Liu Y, Qin W. Mapping cerebral atrophic trajectory from amnestic mild cognitive impairment to Alzheimer's disease. Cereb Cortex 2022; 33:1310-1327. [PMID: 35368064 PMCID: PMC9930625 DOI: 10.1093/cercor/bhac137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/13/2022] [Accepted: 03/13/2022] [Indexed: 11/14/2022] Open
Abstract
Alzheimer's disease (AD) patients suffer progressive cerebral atrophy before dementia onset. However, the region-specific atrophic processes and the influences of age and apolipoprotein E (APOE) on atrophic trajectory are still unclear. By mapping the region-specific nonlinear atrophic trajectory of whole cerebrum from amnestic mild cognitive impairment (aMCI) to AD based on longitudinal structural magnetic resonance imaging data from Alzheimer's disease Neuroimaging Initiative (ADNI) database, we unraveled a quadratic accelerated atrophic trajectory of 68 cerebral regions from aMCI to AD, especially in the superior temporal pole, caudate, and hippocampus. Besides, interaction analyses demonstrated that APOE ε4 carriers had faster atrophic rates than noncarriers in 8 regions, including the caudate, hippocampus, insula, etc.; younger patients progressed faster than older patients in 32 regions, especially for the superior temporal pole, hippocampus, and superior temporal gyrus; and 15 regions demonstrated complex interaction among age, APOE, and disease progression, including the caudate, hippocampus, etc. (P < 0.05/68, Bonferroni correction). Finally, Cox proportional hazards regression model based on the identified region-specific biomarkers could effectively predict the time to AD conversion within 10 years. In summary, cerebral atrophic trajectory mapping could help a comprehensive understanding of AD development and offer potential biomarkers for predicting AD conversion.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoxi He
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hao Ding
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China,School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China
| | - Yu Zhang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yi Ji
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chao Chai
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Meng Liang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China,School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China,Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China,School of Medical Imaging, Tianjin Medical University, Tianjin 300070, China
| | - Yong Liu
- Corresponding author: Wen Qin, Department of Radiology, and Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Anshan Road No 154, Heping District, Tianjin 300052, China. ; Yong Liu, School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China.
| | - Wen Qin
- Corresponding author: Wen Qin, Department of Radiology, and Tianjin Key Lab of Functional Imaging, Tianjin Medical University General Hospital, Anshan Road No 154, Heping District, Tianjin 300052, China. ; Yong Liu, School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China.
| | | |
Collapse
|