1
|
Reus LM, Jansen IE, Tijms BM, Visser PJ, Tesi N, van der Lee SJ, Vermunt L, Peeters CFW, De Groot LA, Hok-A-Hin YS, Chen-Plotkin A, Irwin DJ, Hu WT, Meeter LH, van Swieten JC, Holstege H, Hulsman M, Lemstra AW, Pijnenburg YAL, van der Flier WM, Teunissen CE, del Campo Milan M. Connecting dementia risk loci to the CSF proteome identifies pathophysiological leads for dementia. Brain 2024; 147:3522-3533. [PMID: 38527854 PMCID: PMC11449142 DOI: 10.1093/brain/awae090] [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: 09/04/2023] [Revised: 01/29/2024] [Accepted: 02/23/2024] [Indexed: 03/27/2024] Open
Abstract
Genome-wide association studies have successfully identified many genetic risk loci for dementia, but exact biological mechanisms through which genetic risk factors contribute to dementia remains unclear. Integrating CSF proteomic data with dementia risk loci could reveal intermediate molecular pathways connecting genetic variance to the development of dementia. We tested to what extent effects of known dementia risk loci can be observed in CSF levels of 665 proteins [proximity extension-based (PEA) immunoassays] in a deeply-phenotyped mixed memory clinic cohort [n = 502, mean age (standard deviation, SD) = 64.1 (8.7) years, 181 female (35.4%)], including patients with Alzheimer's disease (AD, n = 213), dementia with Lewy bodies (DLB, n = 50) and frontotemporal dementia (FTD, n = 93), and controls (n = 146). Validation was assessed in independent cohorts (n = 99 PEA platform, n = 198, mass reaction monitoring-targeted mass spectroscopy and multiplex assay). We performed additional analyses stratified according to diagnostic status (AD, DLB, FTD and controls separately), to explore whether associations between CSF proteins and genetic variants were specific to disease or not. We identified four AD risk loci as protein quantitative trait loci (pQTL): CR1-CR2 (rs3818361, P = 1.65 × 10-8), ZCWPW1-PILRB (rs1476679, P = 2.73 × 10-32), CTSH-CTSH (rs3784539, P = 2.88 × 10-24) and HESX1-RETN (rs186108507, P = 8.39 × 10-8), of which the first three pQTLs showed direct replication in the independent cohorts. We identified one AD-specific association between a rare genetic variant of TREM2 and CSF IL6 levels (rs75932628, P = 3.90 × 10-7). DLB risk locus GBA showed positive trans effects on seven inter-related CSF levels in DLB patients only. No pQTLs were identified for FTD loci, either for the total sample as for analyses performed within FTD only. Protein QTL variants were involved in the immune system, highlighting the importance of this system in the pathophysiology of dementia. We further identified pQTLs in stratified analyses for AD and DLB, hinting at disease-specific pQTLs in dementia. Dissecting the contribution of risk loci to neurobiological processes aids in understanding disease mechanisms underlying dementia.
Collapse
Affiliation(s)
- Lianne M Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, CA 90095 CA, USA
| | - Iris E Jansen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Psychiatry, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Niccoló Tesi
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Lisa Vermunt
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Carel F W Peeters
- Mathematical and Statistical Methods group (Biometris), Wageningen University and Research, Wageningen, 6708 PB Wageningen, The Netherlands
| | - Lisa A De Groot
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Rutgers-RWJ Medical School, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08901, USA
| | - Lieke H Meeter
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Henne Holstege
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Marc Hulsman
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Marta del Campo Milan
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, 28003 Madrid, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, 08005 Barcelona, Spain
| |
Collapse
|
2
|
Li QY, Fu Y, Cui XJ, Wang ZT, Tan L. Association of modified dementia risk score with cerebrospinal fluid biomarkers and cognition in adults without dementia. Front Aging Neurosci 2024; 16:1339163. [PMID: 39081396 PMCID: PMC11286572 DOI: 10.3389/fnagi.2024.1339163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction This study aimed to investigate the cognitive profile and prospective cognitive changes in non-demented adults with elevated Modified Dementia Risk Scores (MDRS), while also exploring the potential relationship between these associations and cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) pathology and neuroinflammation. Methods Within the Chinese Alzheimer's Biomarker and LifestylE (CABLE) database, 994 participants without dementia were assessed on MDRS, CSF biomarkers and cognition. We examined the associations of the MDRS with CSF biomarkers and cognitive scores using linear regressions. Causal mediation analyses were conducted to analyze the associations among MDRS, brain pathologies, and cognition. The Alzheimer's Disease Neuroimaging Initiative (ADNI) study was used to validate the mediation effects and to investigate the longitudinal association between MDRS and cognitive decline. Results The results revealed that higher MDRS were linked to poorer cognitive performance (Model 1: PFDR < 0.001; Model 2: PFDR < 0.001) and increases in CSF levels of phosphorylated tau (P-tau, Model 1: PFDR < 0.001; Model 2: PFDR < 0.001), total tau (T-tau, Model 1: PFDR < 0.001; Model 2: PFDR < 0.001), P-tau/Aβ42 ratio (Model 1: PFDR = 0.023; Model 2: PFDR = 0.028), T-tau/Aβ42 ratio (Model 1: PFDR < 0.001; Model 2: PFDR < 0.001) and soluble triggering receptor expressed on myeloid cells 2 (sTrem2, Model 1: PFDR < 0.001; Model 2: PFDR < 0.001) in the CABLE study. The impact of MDRS on cognition was partially mediated by neuroinflammation and tau pathology. These mediation effects were replicated in the ADNI study. Baseline MDRS were significantly associated with future cognitive decline, as indicated by lower scores on the Mini-Mental State Examination (MMSE, Model 1: PFDR = 0.045; Model 2: PFDR < 0.001), ADNI composite memory score (ADNI-MEM, Model 1: PFDR = 0.005; Model 2: PFDR < 0.001), ADNI composite executive function score (ADNI-EF, Model 1: PFDR = 0.045; Model 2: PFDR < 0.001), and higher score on the Alzheimer's Disease Assessment Scale (ADAS13, Model 1: PFDR = 0.045; Model 2: PFDR < 0.001). Discussion The findings of this study revealed significant associations between MDRS and cognitive decline, suggesting a potential role of tau pathology and neuroinflammation in the link between MDRS and poorer cognitive performance in individuals without dementia. Consequently, the MDRS holds promise as a tool for targeted preventive interventions in individuals at high risk of cognitive impairment.
Collapse
Affiliation(s)
- Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xin-Jing Cui
- Department of Outpatient, Qingdao Municipal Hospital, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | | |
Collapse
|
3
|
Wang R, Zhan Y, Zhu W, Yang Q, Pei J. Association of soluble TREM2 with Alzheimer's disease and mild cognitive impairment: a systematic review and meta-analysis. Front Aging Neurosci 2024; 16:1407980. [PMID: 38841103 PMCID: PMC11150578 DOI: 10.3389/fnagi.2024.1407980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Objective Soluble triggering receptor expressed on myeloid cells 2 (sTREM2) is a potential neuroinflammatory biomarker linked to the pathogenesis of Alzheimer's disease (AD) and mild cognitive impairment (MCI). Previous studies have produced inconsistent results regarding sTREM2 levels in various clinical stages of AD. This study aims to establish the correlation between sTREM2 levels and AD progression through a meta-analysis of sTREM2 levels in cerebrospinal fluid (CSF) and blood. Methods Comprehensive searches were conducted in PubMed, Embase, Web of Science, and the Cochrane Library to identify observational studies reporting CSF and blood sTREM2 levels in AD patients, MCI patients, and healthy controls. A random effects meta-analysis was used to calculate the standardized mean difference (SMD) and 95% confidence intervals (CIs). Results Thirty-six observational studies involving 3,016 AD patients, 3,533 MCI patients, and 4,510 healthy controls were included. CSF sTREM2 levels were significantly higher in both the AD [SMD = 0.28, 95% CI (0.15, 0.41)] and MCI groups [SMD = 0.30, 95% CI (0.13, 0.47)] compared to the healthy control group. However, no significant differences in expression were detected between the AD and MCI groups [SMD = 0.09, 95% CI (-0.09, 0.26)]. Furthermore, increased plasma sTREM2 levels were associated with a higher risk of AD [SMD = 0.42, 95% CI (0.01, 0.83)]. Conclusion CSF sTREM2 levels are positively associated with an increased risk of AD and MCI. Plasma sTREM2 levels were notably higher in the AD group than in the control group and may serve as a promising biomarker for diagnosing AD. However, sTREM2 levels are not effective for distinguishing between different disease stages of AD. Further investigations are needed to explore the longitudinal changes in sTREM2 levels, particularly plasma sTREM2 levels, during AD progression. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42024514593.
Collapse
Affiliation(s)
| | | | | | | | - Jian Pei
- Department of Acupuncture, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Hu WT, Kaluzova M, Dawson A, Sotelo V, Papas J, Lemenze A, Shu C, Jomartin M, Nayyar A, Hussain S. Clinical and CSF single-cell profiling of post-COVID-19 cognitive impairment. Cell Rep Med 2024; 5:101561. [PMID: 38744274 PMCID: PMC11148803 DOI: 10.1016/j.xcrm.2024.101561] [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: 08/22/2023] [Revised: 02/15/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Natural history and mechanisms for persistent cognitive symptoms ("brain fog") following acute and often mild COVID-19 are unknown. In a large prospective cohort of people who underwent testing a median of 9 months after acute COVID-19 in the New York City/New Jersey area, we found that cognitive dysfunction is common; is not influenced by mood, fatigue, or sleepiness; and is correlated with MRI changes in very few people. In a subgroup that underwent cerebrospinal fluid analysis, there are no changes related to Alzheimer's disease or neurodegeneration. Single-cell gene expression analysis in the cerebrospinal fluid shows findings consistent with monocyte recruitment, chemokine signaling, cellular stress, and suppressed interferon response-especially in myeloid cells. Longitudinal analysis shows slow recovery accompanied by key alterations in inflammatory genes and increased protein levels of CXCL8, CCL3L1, and sTREM2. These findings suggest that the prognosis for brain fog following COVID-19 correlates with myeloid-related chemokine and interferon-responsive genes.
Collapse
Affiliation(s)
- William T Hu
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA.
| | - Milota Kaluzova
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Alice Dawson
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Victor Sotelo
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Julia Papas
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Alexander Lemenze
- Department of Pathology and Laboratory Medicine, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Carol Shu
- Department of Medicine-Pulmonary and Critical Care, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Mini Jomartin
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Ashima Nayyar
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Sabiha Hussain
- Department of Medicine-Pulmonary and Critical Care, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
5
|
Shue F, White LJ, Hendrix R, Ulrich J, Henson RL, Knight W, Martens YA, Wang N, Roy B, Starling SC, Ren Y, Xiong C, Asmann YW, Syrjanen JA, Vassilaki M, Mielke MM, Timsina J, Sung YJ, Cruchaga C, Holtzman DM, Bu G, Petersen RC, Heckman MG, Kanekiyo T. CSF biomarkers of immune activation and Alzheimer's disease for predicting cognitive impairment risk in the elderly. SCIENCE ADVANCES 2024; 10:eadk3674. [PMID: 38569027 PMCID: PMC10990276 DOI: 10.1126/sciadv.adk3674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/23/2024] [Indexed: 04/05/2024]
Abstract
The immune system substantially influences age-related cognitive decline and Alzheimer's disease (AD) progression, affected by genetic and environmental factors. In a Mayo Clinic Study of Aging cohort, we examined how risk factors like APOE genotype, age, and sex affect inflammatory molecules and AD biomarkers in cerebrospinal fluid (CSF). Among cognitively unimpaired individuals over 65 (N = 298), we measured 365 CSF inflammatory molecules, finding age, sex, and diabetes status predominantly influencing their levels. We observed age-related correlations with AD biomarkers such as total tau, phosphorylated tau-181, neurofilament light chain (NfL), and YKL40. APOE4 was associated with lower Aβ42 and higher SNAP25 in CSF. We explored baseline variables predicting cognitive decline risk, finding age, CSF Aβ42, NfL, and REG4 to be independently correlated. Subjects with older age, lower Aβ42, higher NfL, and higher REG4 at baseline had increased cognitive impairment risk during follow-up. This suggests that assessing CSF inflammatory molecules and AD biomarkers could predict cognitive impairment risk in the elderly.
Collapse
Affiliation(s)
- Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Launia J. White
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rachel Hendrix
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jason Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel L. Henson
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William Knight
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yuka A. Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ni Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Bhaskar Roy
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Yingxue Ren
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 93110, USA
| | - Yan W. Asmann
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jeremy A. Syrjanen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester MN 55905, USA
| | - Maria Vassilaki
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester MN 55905, USA
| | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester MN 55905, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 93110, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 93110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 93110, USA
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Michael G. Heckman
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
6
|
Ackley S, Calmasini C, Bouteloup V, Hill-Jarrett TG, Swinnerton KN, Chêne G, Dufouil C, Glymour MM. Contribution of Global Amyloid-PET Imaging for Predicting Future Cognition in the MEMENTO Cohort. Neurology 2024; 102:e208054. [PMID: 38412412 DOI: 10.1212/wnl.0000000000208054] [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: 03/01/2023] [Accepted: 10/16/2023] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Global amyloid-PET is associated with cognition and cognitive decline, but most research on this association does not account for past cognitive information. We assessed the prognostic benefit of amyloid-PET measures for future cognition when prior cognitive assessments are available, evaluating the added value of amyloid measures beyond information on multiple past cognitive assessments. METHODS The French MEMENTO cohort (a cohort of outpatients from French research memory centers to improve knowledge on Alzheimer disease and related disorders) includes older outpatients with incipient cognitive changes, but no dementia diagnosis at inclusion. Global amyloid burden was assessed using positron emission tomography (amyloid-PET) for a subset of participants; semiannual cognitive testing was subsequently performed. We predicted mini-mental state examination (MMSE) scores using demographic characteristics (age, sex, marital status, and education) alone or in combination with information on prior cognitive measures. The added value of amyloid burden as a predictor in these models was evaluated with percent reduction of the mean squared error (MSE). All models were conducted separately for evaluating the added value of dichotomous amyloid positivity status compared with a continuous amyloid-standardized uptake-value ratio. RESULTS Our analytic sample comprised 510 individuals who underwent amyloid-PET scans with at least 4 MMSE assessments. The mean age at the PET scan was 71.6 (standard deviation 7.4) years; 60.7% were female. The median follow-up was 4.6 years (interquartile range: 0.9 years). Adding amyloid burden when adjusting for only demographic characteristics reduced the MSE of predictions by 5.08% (95% CI 0.97%-10.86%) and 12.64% (95% CI 3.35%-25.28%) for binary and continuous amyloid, respectively. If the model included 1 past MMSE measure, the MSE improvement was 3.51% (95% CI 1.01%-7.28%) when adding binary amyloid and 8.83% (95% CI 2.63%-16.37%) when adding continuous amyloid. Improvements in model fit were smaller with the addition of amyloid burden when more than 1 past cognitive assessment was included. For all models incorporating past cognitive assessments, differences in predictions amounted to a fraction of 1 MMSE point on average. DISCUSSION In a clinical setting, global amyloid burden did not appreciably improve cognitive predictions when past cognitive assessments were available. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT02164643.
Collapse
Affiliation(s)
- Sarah Ackley
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Camilla Calmasini
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Vincent Bouteloup
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Tanisha G Hill-Jarrett
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Kaitlin N Swinnerton
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Geneviève Chêne
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Carole Dufouil
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - M M Glymour
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| |
Collapse
|
7
|
Veitch DP, Weiner MW, Miller M, Aisen PS, Ashford MA, Beckett LA, Green RC, Harvey D, Jack CR, Jagust W, Landau SM, Morris JC, Nho KT, Nosheny R, Okonkwo O, Perrin RJ, Petersen RC, Rivera Mindt M, Saykin A, Shaw LM, Toga AW, Tosun D. The Alzheimer's Disease Neuroimaging Initiative in the era of Alzheimer's disease treatment: A review of ADNI studies from 2021 to 2022. Alzheimers Dement 2024; 20:652-694. [PMID: 37698424 PMCID: PMC10841343 DOI: 10.1002/alz.13449] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) aims to improve Alzheimer's disease (AD) clinical trials. Since 2006, ADNI has shared clinical, neuroimaging, and cognitive data, and biofluid samples. We used conventional search methods to identify 1459 publications from 2021 to 2022 using ADNI data/samples and reviewed 291 impactful studies. This review details how ADNI studies improved disease progression understanding and clinical trial efficiency. Advances in subject selection, detection of treatment effects, harmonization, and modeling improved clinical trials and plasma biomarkers like phosphorylated tau showed promise for clinical use. Biomarkers of amyloid beta, tau, neurodegeneration, inflammation, and others were prognostic with individualized prediction algorithms available online. Studies supported the amyloid cascade, emphasized the importance of neuroinflammation, and detailed widespread heterogeneity in disease, linked to genetic and vascular risk, co-pathologies, sex, and resilience. Biological subtypes were consistently observed. Generalizability of ADNI results is limited by lack of cohort diversity, an issue ADNI-4 aims to address by enrolling a diverse cohort.
Collapse
Affiliation(s)
- Dallas P. Veitch
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Michael W. Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Melanie Miller
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Miriam A. Ashford
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
| | - Laurel A. Beckett
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Robert C. Green
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalBroad Institute Ariadne Labs and Harvard Medical SchoolBostonMassachusettsUSA
| | - Danielle Harvey
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | | - William Jagust
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Susan M. Landau
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - John C. Morris
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Kwangsik T. Nho
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rachel Nosheny
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ozioma Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Richard J. Perrin
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Monica Rivera Mindt
- Department of PsychologyLatin American and Latino Studies InstituteAfrican and African American StudiesFordham UniversityNew YorkNew YorkUSA
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Andrew Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine and the PENN Alzheimer's Disease Research CenterCenter for Neurodegenerative ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arthur W. Toga
- Laboratory of Neuro ImagingInstitute of Neuroimaging and InformaticsKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Duygu Tosun
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | |
Collapse
|
8
|
Schlepckow K, Morenas-Rodríguez E, Hong S, Haass C. Stimulation of TREM2 with agonistic antibodies-an emerging therapeutic option for Alzheimer's disease. Lancet Neurol 2023; 22:1048-1060. [PMID: 37863592 DOI: 10.1016/s1474-4422(23)00247-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 10/22/2023]
Abstract
Neurodegenerative disorders, including Alzheimer's disease, are associated with microgliosis. Microglia have long been considered to have detrimental roles in Alzheimer's disease. However, functional analyses of genes encoding risk factors that are linked to late-onset Alzheimer's disease, and that are enriched or exclusively expressed in microglia, have revealed unexpected protective functions. One of the major risk genes for Alzheimer's disease is TREM2. Risk variants of TREM2 are loss-of-function mutations affecting chemotaxis, phagocytosis, lipid and energy metabolism, and survival and proliferation. Agonistic anti-TREM2 antibodies have been developed to boost these protective functions in patients with intact TREM2 alleles. Several anti-TREM2 antibodies are in early clinical trials, and current efforts aim to achieve more efficient transport of these antibodies across the blood-brain barrier. PET imaging could be used to monitor target engagement. Data from animal models, and biomarker studies in patients, further support a rationale for boosting TREM2 functions during the preclinical stage of Alzheimer's disease.
Collapse
Affiliation(s)
- Kai Schlepckow
- German Centre for Neurodegenerative Diseases, Munich, Germany
| | - Estrella Morenas-Rodríguez
- Memory Unit, Department of Neurology, Hospital Universitario 12 de Octubre, Madrid, Spain; Group of Neurogenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Christian Haass
- German Centre for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Centre, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
9
|
Dai Y, Cheng Y, Zhou Z, Li Z, Luo Y, Qiu H. A contrast-enhanced CT-based whole-spleen radiomics signature for early prediction of oxaliplatin-related thrombocytopenia in patients with gastrointestinal malignancies: a retrospective study. PeerJ 2023; 11:e16230. [PMID: 37849829 PMCID: PMC10578303 DOI: 10.7717/peerj.16230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/12/2023] [Indexed: 10/19/2023] Open
Abstract
Background Thrombocytopenia is a common adverse event of oxaliplatin-based chemotherapy. Grade 2 or higher oxaliplatin-related thrombocytopenia may result in dose reduction, discontinuation or delay initiation of chemotherapy and may adversely affect the therapeutic efficacy and even overall survival of patients. Early recognition of patients at risk of developing grade 2 or higher thrombocytopenia is critical. However, to date there is no well-established method to early identify patients at high risk. The aims of this study were to develop and validate a contrast-enhanced CT-based whole-spleen radiomics signature for early prediction of grade 2 or higher thrombocytopenia in patients with gastrointestinal malignancies treated with oxaliplatin-based chemotherapy and to explore the incremental value of combining the radiomics signature and conventional clinical factors for risk prediction. Methods A total of 119 patients with gastrointestinal malignancies receiving oxaliplatin-based chemotherapy from March 2017 to December 2020 were retrospectively included and randomly divided into a training cohort (n = 85) and a validation cohort (n = 34). Grade 2 or higher thrombocytopenia occurred in 26.1% of patients (22 and nine patients in the training and validation cohort, respectively) with a median time interval of 101 days from the start of chemotherapy. The whole-spleen radiomics features were extracted on the portal venous phase of the first follow-up CT images. The least absolute shrinkage and selection operator (LASSO) algorithm was applied to select radiomics features and to build the radiomics signature for the prediction of grade 2 or higher thrombocytopenia. A clinical model that included clinical factors only and a clinical-radiomics model that incorporated clinical factors and radiomics signature were constructed. The performances of both models were evaluated and compared in the training, validation and the whole cohorts. Results The radiomics signature yielded favorable performance in predicting grade 2 or higher thrombocytopenia, with the area under the curve (AUC), sensitivity and specificity being 0.865, 81.8% and 84.1% in the training cohort and 0.747, 77.8% and 80.0% in the validation cohort. The AUCs of the clinical-radiomics model in the training and validation cohorts reached 0.913 (95% CI [0.720-0.935]) and 0.867 (95% CI [0.727-1.000]), greater than the AUCs of the clinical model. Integrated discrimination improvement (IDI) index showed that incorporating radiomic signature into conventional clinical factors significantly improved the predictive accuracy by 17.0% (95% CI [4.9%-29.1%], p = 0.006) in the whole cohort. Conclusions Contrast-enhanced CT-based whole-spleen radiomics signature might serve as an early predictor for grade 2 or higher thrombocytopenia during oxaliplatin-based chemotherapy in patients with gastrointestinal malignancies and provide incremental value over conventional clinical factors.
Collapse
Affiliation(s)
- Yuhong Dai
- Department of Oncology, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiqi Cheng
- Department of Radiology, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziling Zhou
- Department of Radiology, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Li
- Department of Radiology, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Luo
- Department of Radiology, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Qiu
- Department of Oncology, Tongji hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Yang X, Wen J, Yang H, Jones IR, Zhu X, Liu W, Li B, Clelland CD, Luo W, Wong MY, Ren X, Cui X, Song M, Liu H, Chen C, Eng N, Ravichandran M, Sun Y, Lee D, Van Buren E, Jiang MZ, Chan CSY, Ye CJ, Perera RM, Gan L, Li Y, Shen Y. Functional characterization of Alzheimer's disease genetic variants in microglia. Nat Genet 2023; 55:1735-1744. [PMID: 37735198 PMCID: PMC10939305 DOI: 10.1038/s41588-023-01506-8] [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: 07/12/2022] [Accepted: 08/20/2023] [Indexed: 09/23/2023]
Abstract
Candidate cis-regulatory elements (cCREs) in microglia demonstrate the most substantial enrichment for Alzheimer's disease (AD) heritability compared to other brain cell types. However, whether and how these genome-wide association studies (GWAS) variants contribute to AD remain elusive. Here we prioritize 308 previously unreported AD risk variants at 181 cCREs by integrating genetic information with microglia-specific 3D epigenome annotation. We further establish the link between functional variants and target genes by single-cell CRISPRi screening in microglia. In addition, we show that AD variants exhibit allelic imbalance on target gene expression. In particular, rs7922621 is the effective variant in controlling TSPAN14 expression among other nominated variants in the same cCRE and exerts multiple physiological effects including reduced cell surface ADAM10 and altered soluble TREM2 (sTREM2) shedding. Our work represents a systematic approach to prioritize and characterize AD-associated variants and provides a roadmap for advancing genetic association to experimentally validated cell-type-specific phenotypes and mechanisms.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Jia Wen
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Han Yang
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Ian R Jones
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Xiaodong Zhu
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York City, NY, USA
| | - Weifang Liu
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Bingkun Li
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Claire D Clelland
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York City, NY, USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York City, NY, USA
| | - Xingjie Ren
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Xiekui Cui
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Michael Song
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Hongjiang Liu
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Cady Chen
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Nicolas Eng
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Yang Sun
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - David Lee
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eric Van Buren
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Min-Zhi Jiang
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Candace S Y Chan
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Chun Jimmie Ye
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Rushika M Perera
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York City, NY, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA.
- Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA.
| | - Yin Shen
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
11
|
Giannisis A, Al-Grety A, Carlsson H, Howell JC, Hu WT, Kultima K, Nielsen HM. Plasma apolipoprotein E levels, isoform composition, and dimer profile in relation to plasma lipids in racially diverse patients with Alzheimer's disease and mild cognitive impairment. Alzheimers Res Ther 2023; 15:119. [PMID: 37400888 PMCID: PMC10316569 DOI: 10.1186/s13195-023-01262-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND The APOEε4-promoted risk of Alzheimer's disease (AD) is lower in Black/African-Americans (B/AAs), compared to non-Hispanic whites (NHWs). Previous studies reported lower plasma apolipoprotein E (apoE) levels in NHW APOEε4-carriers compared to non-carriers, and low plasma apoE levels were directly associated with an increased risk of AD and all dementia. We further showed that APOEε3/ε3 AD patients exhibited reduced plasma apoE dimers compared to corresponding control subjects. Whether plasma apoE levels and apoE dimer formation differ between races/ethnicities and therefore may help explain AD risk racial disparity remains to be elucidated. METHODS Using mass spectrometry, we determined total plasma apoE and apoE isoform levels in a cohort of B/AAs (n = 58) and NHWs (n = 67) including subjects with normal cognition (B/AA: n = 25, NHW: n = 28), mild cognitive impairment (MCI) (B/AA: n = 24, NHW: n = 24), or AD dementia (B/AA: n = 9, NHW: n = 15). Additionally, we used non-reducing western blot analysis to assess the distribution of plasma apoE into monomers/disulfide-linked dimers. Plasma total apoE, apoE isoform levels, and % apoE monomers/dimers were assessed for correlations with cognition, cerebrospinal fluid (CSF) AD biomarkers, sTREM2, neurofilament light protein (NfL), and plasma lipids. RESULTS Plasma apoE was predominantly monomeric in both racial groups and the monomer/dimer distribution was not affected by disease status, or correlated with CSF AD biomarkers, but associated with plasma lipids. Plasma total apoE levels were not related to disease status and only in the NHW subjects we observed lower plasma apoE levels in the APOEε4/ε4-carriers. Total plasma apoE levels were 13% higher in B/AA compared to NHW APOEε4/ε4 subjects and associated with plasma high-density lipoprotein (HDL) in NHW subjects but with low-density lipoprotein levels (LDL) in the B/AA subjects. Higher plasma apoE4 levels, exclusively in APOEε3/ε4 B/AA subjects, were linked to higher plasma total cholesterol and LDL levels. In the controls, NHWs and B/AAs exhibited opposite associations between plasma apoE and CSF t-tau. CONCLUSIONS The previously reported lower APOEε4-promoted risk of AD in B/AA subjects may be associated with differences in plasma apoE levels and lipoprotein association. Whether differences in plasma apoE levels between races/ethnicities result from altered APOEε4 expression or turnover, needs further elucidation.
Collapse
Affiliation(s)
- Andreas Giannisis
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius Väg 16B, 114 18, Stockholm, Sweden
| | - Asma Al-Grety
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Henrik Carlsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | | | - William T Hu
- Department of Neurology, Emory University, Atlanta, GA, USA
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, and Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius Väg 16B, 114 18, Stockholm, Sweden.
| |
Collapse
|
12
|
Hu WT, Nayyar A, Kaluzova M. Charting the Next Road Map for CSF Biomarkers in Alzheimer's Disease and Related Dementias. Neurotherapeutics 2023; 20:955-974. [PMID: 37378862 PMCID: PMC10457281 DOI: 10.1007/s13311-023-01370-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 06/29/2023] Open
Abstract
Clinical prediction of underlying pathologic substrates in people with Alzheimer's disease (AD) dementia or related dementia syndromes (ADRD) has limited accuracy. Etiologic biomarkers - including cerebrospinal fluid (CSF) levels of AD proteins and cerebral amyloid PET imaging - have greatly modernized disease-modifying clinical trials in AD, but their integration into medical practice has been slow. Beyond core CSF AD biomarkers (including beta-amyloid 1-42, total tau, and tau phosphorylated at threonine 181), novel biomarkers have been interrogated in single- and multi-centered studies with uneven rigor. Here, we review early expectations for ideal AD/ADRD biomarkers, assess these goals' future applicability, and propose study designs and performance thresholds for meeting these ideals with a focus on CSF biomarkers. We further propose three new characteristics: equity (oversampling of diverse populations in the design and testing of biomarkers), access (reasonable availability to 80% of people at risk for disease, along with pre- and post-biomarker processes), and reliability (thorough evaluation of pre-analytical and analytical factors influencing measurements and performance). Finally, we urge biomarker scientists to balance the desire and evidence for a biomarker to reflect its namesake function, indulge data- as well as theory-driven associations, re-visit the subset of rigorously measured CSF biomarkers in large datasets (such as Alzheimer's disease neuroimaging initiative), and resist the temptation to favor ease over fail-safe in the development phase. This shift from discovery to application, and from suspended disbelief to cogent ingenuity, should allow the AD/ADRD biomarker field to live up to its billing during the next phase of neurodegenerative disease research.
Collapse
Affiliation(s)
- William T Hu
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA.
- Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Ashima Nayyar
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA
| | - Milota Kaluzova
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA
| |
Collapse
|
13
|
Salai KHT, Wu LY, Chong JR, Chai YL, Gyanwali B, Robert C, Hilal S, Venketasubramanian N, Dawe GS, Chen CP, Lai MKP. Elevated Soluble TNF-Receptor 1 in the Serum of Predementia Subjects with Cerebral Small Vessel Disease. Biomolecules 2023; 13:biom13030525. [PMID: 36979460 PMCID: PMC10046240 DOI: 10.3390/biom13030525] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023] Open
Abstract
Tumor necrosis factor-receptor 1 (TNF-R1)-mediated signaling is critical to the regulation of inflammatory responses. TNF-R1 can be proteolytically released into systemic blood circulation in a soluble form (sTNF-R1), where it binds to circulating TNF and functions to attenuate TNF-mediated inflammation. Increases of peripheral sTNF-R1 have been reported in both Alzheimer’s disease (AD) dementia and vascular dementia (VaD). However, the status of sTNF-R1 in predementia subjects (cognitive impairment, no dementia, CIND) is unknown, and putative associations with cerebral small vessel disease (CSVD), as well as with longitudinal changes in cognitive functions are unclear. We measured baseline serum sTNF-R1 in a longitudinally assessed cohort of 93 controls and 103 CIND, along with neuropsychological evaluations and neuroimaging assessments. Serum sTNF-R1 levels were increased in CIND compared with controls (p < 0.001). Higher baseline sTNF-R1 levels were specifically associated with lacunar infarcts (rate ratio = 6.91, 95% CI 3.19–14.96, p < 0.001), as well as lower rates of cognitive decline in the CIND subgroup. Our data suggest that sTNF-R1 interacts with vascular cognitive impairment in a complex manner at predementia stages, with elevated levels associated with more severe CSVD at baseline, but which may subsequently be protective against cognitive decline.
Collapse
Affiliation(s)
- Kaung H. T. Salai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Liu-Yun Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore 117600, Singapore
| | - Joyce R. Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore 117600, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore 117600, Singapore
| | - Bibek Gyanwali
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore 117600, Singapore
| | - Caroline Robert
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore 117600, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117597, Singapore
- Departments of Epidemiology and Radiology & Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | | | - Gavin S. Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Neurobiology Programme, Life Sciences Institute, Centre for Life Sciences, National University of Singapore, Singapore 117456, Singapore
| | - Christopher P. Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore 117600, Singapore
| | - Mitchell K. P. Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore 117600, Singapore
- Correspondence:
| |
Collapse
|
14
|
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: 8.0] [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
|
15
|
Schein CH. Distinguishing Curable from Progressive Dementias for Defining Cancer Care Options. Cancers (Basel) 2023; 15:cancers15041055. [PMID: 36831398 PMCID: PMC9954275 DOI: 10.3390/cancers15041055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/06/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
The likelihood of a diagnosis of dementia increases with a person's age, as is also the case for many cancers, including melanoma and multiple myeloma, where the median age of diagnosis is above 60 years. However, patients diagnosed with dementia are less likely to be offered invasive curative therapies for cancer. Together with analysis of diet and medication history, advanced imaging methods and genetic profiling can now indicate more about syndromes causing the neurological symptoms. Cachexia, malnutrition, dehydration, alcohol consumption, and even loneliness can all accentuate or cause the "3Ds" of dementia, delirium and depression. Many common drugs, especially in the context of polypharmacy, can cause cognitive difficulties resembling neurodegenerative disease. These syndromes may be reversed by diet, social and caregiver changes, and stopping potentially inappropriate medications (PIMs). More insidious are immune reactions to many different autoantigens, some of which are related to cancers and tumors. These can induce movement and cognitive difficulties that mimic Alzheimer's and Parkinson's diseases and other ataxias associated with aging. Paraneoplastic neurological syndromes may be reversed by directed immunotherapies if detected in their early stages but are best treated by removal of the causative tumor. A full genetic workup should be done for all individuals as soon as possible after diagnosis, to guide less invasive treatments suitable for frail individuals. While surgical interventions may be contraindicated, genetic profile guided immunotherapies, oral treatments, and radiation may be equally curative in a significant number of cancers.
Collapse
Affiliation(s)
- Catherine H Schein
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| |
Collapse
|
16
|
Verma S, Perry K, Razdan R, Howell JC, Dawson AL, Hu WT. CSF IL-8 Associated with Response to Gene Therapy in a Case Series of Spinal Muscular Atrophy. Neurotherapeutics 2023; 20:245-253. [PMID: 36289175 PMCID: PMC9607706 DOI: 10.1007/s13311-022-01305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2022] [Indexed: 11/27/2022] Open
Abstract
Gene therapies have greatly changed the outlook in spinal muscular atrophy (SMA), and this disorder provides a rare opportunity to study longitudinal biomarker changes correlated with reduced disease burden and improved clinical outcomes. Recent work suggests clinical response to correlate with declining cerebrospinal fluid (CSF) levels of the neurodegenerative marker neurofilament light chain (NfL) in children receiving serial anti-sense oligonucleotide therapy. However, change in CSF NfL levels is no longer a practical biomarker as more children undergo single-dose gene replacement therapy. Here we leverage serial CSF samples (median of 4 per child) collected in 13 children with SMA undergoing anti-sense oligonucleotide therapy to characterize the longitudinal profiles of NfL as well as inflammatory and neuronal proteins. In contrast to neurodegeneration in adults, we found NfL levels to first decrease following initiation of treatment but then increase upon further treatment and improved motor functions. We then examined additional CSF inflammatory and neuronal markers for linear association with motor function during SMA treatment. We identified longitudinal IL-8 levels to inversely correlate with motor functions determined by clinical examination (F(1, 47) = 12.903, p = 0.001) or electromyography in the abductor pollicis brevis muscle (p = 0.064). In keeping with this, lower baseline IL-8 levels were associated with better longitudinal outcomes, even though this difference diminished over 2 years in the younger group. We thus propose CSF IL-8 as a biomarker for baseline function and short-term treatment response in SMA, and a candidate biomarker for future treatment trials in other neurodegenerative disorders.
Collapse
Affiliation(s)
- Sumit Verma
- Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, 30324, USA
- Department of Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, 30324, USA
- Department of Neurosciences, Children's Healthcare of Atlanta, Atlanta, GA, 30324, USA
| | - Kelsey Perry
- Department of Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, 30324, USA
| | - Raj Razdan
- Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, 30324, USA
| | - J Christina Howell
- Department of Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, 30324, USA
| | - Alice L Dawson
- Department of Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, 30324, USA
- Health Care Policy, and Aging Research, Rutgers-Robert Wood Johnson Medical School and Rutgers Institute for Health, New Brunswick, NJ, 08901, USA
- Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, 30324, USA.
- Health Care Policy, and Aging Research, Rutgers-Robert Wood Johnson Medical School and Rutgers Institute for Health, New Brunswick, NJ, 08901, USA.
- Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
17
|
Combination of Serum and Plasma Biomarkers Could Improve Prediction Performance for Alzheimer’s Disease. Genes (Basel) 2022; 13:genes13101738. [PMID: 36292623 PMCID: PMC9601501 DOI: 10.3390/genes13101738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) can be predicted either by serum or plasma biomarkers, and a combination may increase predictive power, but due to the high complexity of machine learning, it may also incur overfitting problems. In this paper, we investigated whether combining serum and plasma biomarkers with feature selection could improve prediction performance for AD. 150 D patients and 150 normal controls (NCs) were enrolled for a serum test, and 100 patients and 100 NCs were enrolled for the plasma test. Among these, 79 ADs and 65 NCs had serum and plasma samples in common. A 10 times repeated 5-fold cross-validation model and a feature selection method were used to overcome the overfitting problem when serum and plasma biomarkers were combined. First, we tested to see if simply adding serum and plasma biomarkers improved prediction performance but also caused overfitting. Then we employed a feature selection algorithm we developed to overcome the overfitting problem. Lastly, we tested the prediction performance in a 10 times repeated 5-fold cross validation model for training and testing sets. We found that the combined biomarkers improved AD prediction but also caused overfitting. A further feature selection based on the combination of serum and plasma biomarkers solved the problem and produced an even higher prediction performance than either serum or plasma biomarkers on their own. The combined feature-selected serum–plasma biomarkers may have critical implications for understanding the pathophysiology of AD and for developing preventative treatments.
Collapse
|
18
|
Taddei RN, Sanchez-Mico MV, Bonnar O, Connors T, Gaona A, Denbow D, Frosch MP, Gómez-Isla T. Changes in glial cell phenotypes precede overt neurofibrillary tangle formation, correlate with markers of cortical cell damage, and predict cognitive status of individuals at Braak III-IV stages. Acta Neuropathol Commun 2022; 10:72. [PMID: 35534858 PMCID: PMC9082857 DOI: 10.1186/s40478-022-01370-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 11/10/2022] Open
Abstract
Clinico-pathological correlation studies show that some otherwise healthy elderly individuals who never developed cognitive impairment harbor a burden of Alzheimer's disease lesions (plaques and tangles) that would be expected to result in dementia. In the absence of comorbidities explaining such discrepancies, there is a need to identify other brain changes that meaningfully contribute to the cognitive status of an individual in the face of such burdens of plaques and tangles. Glial inflammatory responses, a universal phenomenon in symptomatic AD, show robust association with degree of cognitive impairment, but their significance in early tau pathology stages and contribution to the trajectory of cognitive decline at an individual level remain widely unexplored. We studied 55 brains from individuals at intermediate stages of tau tangle pathology (Braak III-IV) with diverging antemortem cognition (demented vs. non-demented, here termed `resilient'), and age-matched cognitively normal controls (Braak 0-II). We conducted quantitative assessments of amyloid and tau lesions, cellular vulnerability markers, and glial phenotypes in temporal pole (Braak III-IV region) and visual cortex (Braak V-VI region) using artificial-intelligence based semiautomated quantifications. We found distinct glial responses with increased proinflammatory and decreased homeostatic markers, both in regions with tau tangles (temporal pole) and without overt tau deposits (visual cortex) in demented but not in resilient. These changes were significantly associated with markers of cortical cell damage. Similar phenotypic glial changes were detected in the white matter of demented but not resilient and were associated with higher burden of overlying cortical cellular damage in regions with and without tangles. Our data suggest that changes in glial phenotypes in cortical and subcortical regions represent an early phenomenon that precedes overt tau deposition and likely contributes to cell damage and loss of brain function predicting the cognitive status of individuals at intermediate stages of tau aggregate burden (Braak III-IV).
Collapse
Affiliation(s)
- Raquel N Taddei
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- Department of Neurology, Dementia Research Institute, University College London, London, UK
| | - Maria V Sanchez-Mico
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Orla Bonnar
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Theresa Connors
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Angelica Gaona
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Dominique Denbow
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Matthew P Frosch
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gómez-Isla
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA.
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA.
| |
Collapse
|
19
|
Pomilio AB, Vitale AA, Lazarowski AJ. Neuroproteomics Chip-Based Mass Spectrometry and Other Techniques for Alzheimer´S Disease Biomarkers – Update. Curr Pharm Des 2022; 28:1124-1151. [DOI: 10.2174/1381612828666220413094918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer's disease (AD) is a progressive neurodegenerative disease of growing interest given that there is cognitive damage and symptom onset acceleration. Therefore, it is important to find AD biomarkers for early diagnosis, disease progression, and discrimination of AD and other diseases.
Objective:
To update the relevance of mass spectrometry for the identification of peptides and proteins involved in AD useful as discriminating biomarkers.
Methods:
Proteomics and peptidomics technologies that show the highest possible specificity and selectivity for AD biomarkers are analyzed, together with the biological fluids used. In addition to positron emission tomography and magnetic resonance imaging, MALDI-TOF mass spectrometry is widely used to identify proteins and peptides involved in AD. The use of protein chips in SELDI technology and electroblotting chips for peptides makes feasible small amounts (L) of samples for analysis.
Results:
Suitable biomarkers are related to AD pathology, such as intracellular neurofibrillary tangles; extraneuronal senile plaques; neuronal and axonal degeneration; inflammation and oxidative stress. Recently, peptides were added to the candidate list, which are not amyloid-b or tau fragments, but are related to coagulation, brain plasticity, and complement/neuroinflammation systems involving the neurovascular unit.
Conclusion:
The progress made in the application of mass spectrometry and recent chip techniques is promising for discriminating between AD, mild cognitive impairment, and matched healthy controls. The application of this technique to blood samples from patients with AD has shown to be less invasive and fast enough to determine the diagnosis, stage of the disease, prognosis, and follow-up of the therapeutic response.
Collapse
Affiliation(s)
- Alicia B. Pomilio
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Arturo A. Vitale
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Alberto J. Lazarowski
- Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Córdoba 2351, C1120AAF Buenos Aires, Argentina
| |
Collapse
|
20
|
Chowdhury A, Witte S, Aich A. Role of Mitochondrial Nucleic Acid Sensing Pathways in Health and Patho-Physiology. Front Cell Dev Biol 2022; 10:796066. [PMID: 35223833 PMCID: PMC8873532 DOI: 10.3389/fcell.2022.796066] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/14/2022] [Indexed: 12/23/2022] Open
Abstract
Mitochondria, in symbiosis with the host cell, carry out a wide variety of functions from generating energy, regulating the metabolic processes, cell death to inflammation. The most prominent function of mitochondria relies on the oxidative phosphorylation (OXPHOS) system. OXPHOS heavily influences the mitochondrial-nuclear communication through a plethora of interconnected signaling pathways. Additionally, owing to the bacterial ancestry, mitochondria also harbor a large number of Damage Associated Molecular Patterns (DAMPs). These molecules relay the information about the state of the mitochondrial health and dysfunction to the innate immune system. Consequently, depending on the intracellular or extracellular nature of detection, different inflammatory pathways are elicited. One group of DAMPs, the mitochondrial nucleic acids, hijack the antiviral DNA or RNA sensing mechanisms such as the cGAS/STING and RIG-1/MAVS pathways. A pro-inflammatory response is invoked by these signals predominantly through type I interferon (T1-IFN) cytokines. This affects a wide range of organ systems which exhibit clinical presentations of auto-immune disorders. Interestingly, tumor cells too, have devised ingenious ways to use the mitochondrial DNA mediated cGAS-STING-IRF3 response to promote neoplastic transformations and develop tumor micro-environments. Thus, mitochondrial nucleic acid-sensing pathways are fundamental in understanding the source and nature of disease initiation and development. Apart from the pathological interest, recent studies also attempt to delineate the structural considerations for the release of nucleic acids across the mitochondrial membranes. Hence, this review presents a comprehensive overview of the different aspects of mitochondrial nucleic acid-sensing. It attempts to summarize the nature of the molecular patterns involved, their release and recognition in the cytoplasm and signaling. Finally, a major emphasis is given to elaborate the resulting patho-physiologies.
Collapse
Affiliation(s)
- Arpita Chowdhury
- Department of Cellular Biochemistry, University Medical Center, Göttingen, Germany
| | - Steffen Witte
- Department of Cellular Biochemistry, University Medical Center, Göttingen, Germany
| | - Abhishek Aich
- Department of Cellular Biochemistry, University Medical Center, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging, from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
21
|
Jeremic D, Jiménez-Díaz L, Navarro-López JD. Past, present and future of therapeutic strategies against amyloid-β peptides in Alzheimer's disease: a systematic review. Ageing Res Rev 2021; 72:101496. [PMID: 34687956 DOI: 10.1016/j.arr.2021.101496] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease in ageing, affecting around 46 million people worldwide but few treatments are currently available. The etiology of AD is still puzzling, and new drugs development and clinical trials have high failure rates. Urgent outline of an integral (multi-target) and effective treatment of AD is needed. Accumulation of amyloid-β (Aβ) peptides is considered one of the fundamental neuropathological pillars of the disease, and its dyshomeostasis has shown a crucial role in AD onset. Therefore, many amyloid-targeted therapies have been investigated. Here, we will systematically review recent (from 2014) investigational, follow-up and review studies focused on anti-amyloid strategies to summarize and analyze their current clinical potential. Combination of anti-Aβ therapies with new developing early detection biomarkers and other therapeutic agents acting on early functional AD changes will be highlighted in this review. Near-term approval seems likely for several drugs acting against Aβ, with recent FDA approval of a monoclonal anti-Aβ oligomers antibody -aducanumab- raising hopes and controversies. We conclude that, development of oligomer-epitope specific Aβ treatment and implementation of multiple improved biomarkers and risk prediction methods allowing early detection, together with therapies acting on other factors such as hyperexcitability in early AD, could be the key to slowing this global pandemic.
Collapse
|
22
|
Tsai HH, Chen YF, Yen RF, Lo YL, Yang KC, Jeng JS, Tsai LK, Chang CF. Plasma soluble TREM2 is associated with white matter lesions independent of amyloid and tau. Brain 2021; 144:3371-3380. [PMID: 34515756 DOI: 10.1093/brain/awab332] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/21/2021] [Accepted: 08/08/2021] [Indexed: 12/20/2022] Open
Abstract
Cerebral small vessel disease is one of the most common causes of cognitive decline and stroke. While several lines of evidence have established a relationship between inflammation and cerebrovascular pathology, the mechanistic link has not yet been elucidated. Recent studies suggest activation of immune mediators, including the soluble form of triggering receptor expressed on myeloid cells 2 (TREM2), may be critical regulators. In this study, we compared the plasma levels of soluble TREM2 and its correlations with neuroimaging markers and cerebral amyloid load in ten patients with Alzheimer's disease and 66 survivors of spontaneous intracerebral haemorrhage with cerebral amyloid angiopathy or hypertensive small vessel disease, two of the most common types of sporadic small vessel disease. We performed brain MRI and 11C-Pittsburgh compound B PET for all participants to evaluate radiological small vessel disease markers and cerebral amyloid burden, and 18F-T807 PET in a subgroup of patients to evaluate cortical tau pathology. Plasma soluble TREM2 levels were comparable between patients with Alzheimer's disease and small vessel disease (P=0.690). In patients with small vessel disease, plasma soluble TREM2 was significantly associated with white matter hyperintensity volume (P<0.001), but not with cerebral amyloid load. Among patients with Alzheimer's disease and cerebral amyloid angiopathy, plasma soluble TREM2 was independently associated with a tau-positive scan (P=0.001) and white matter hyperintensity volume (P=0.013), but not amyloid load (P=0.221). Our results indicate plasma soluble TREM2 is associated with white matter hyperintensity independent of amyloid and tau pathology. These findings highlight the potential utility of plasma soluble TREM2 as a strong predictive marker for small vessel disease-related white matter injury and hold clinical implications for targeting the innate immune response when treating this disease.
Collapse
Affiliation(s)
- Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan.,Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Fang Chen
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yen-Ling Lo
- Department of Neurology, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan
| | - Kai-Chien Yang
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jiann-Shing Jeng
- Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Che-Feng Chang
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|