1
|
Fleck L, Buss C, Bauer M, Stein M, Mekle R, Kock L, Klawitter H, Godara M, Ramler J, Entringer S, Endres M, Heim C. Early-Life Adversity Predicts Markers of Aging-Related Neuroinflammation, Neurodegeneration, and Cognitive Impairment in Women. Ann Neurol 2025; 97:642-656. [PMID: 39786167 PMCID: PMC11889533 DOI: 10.1002/ana.27161] [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: 07/05/2024] [Revised: 11/24/2024] [Accepted: 11/29/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVE Despite the overwhelming evidence for profound and longstanding effects of early-life stress (ELS) on inflammation, brain structure, and molecular aging, its impact on human brain aging and risk for neurodegenerative disease is poorly understood. We examined the impact of ELS severity in interaction with age on blood-based markers of neuroinflammation and neurodegeneration, brain volumes, and cognitive function in middle-aged women. METHODS We recruited 179 women (aged 30-60 years) with and without ELS exposure before the onset of puberty. Using Simoa technology, we assessed blood-based markers of neuroinflammation and neurodegeneration, including serum concentrations of glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL). We further obtained T1-weighted and T2-weighted magnetic resonance images to assess brain volumes and we assessed cognitive performance sensitive to early impairments associated with the development of dementia, using the Cambridge Neuropsychological Automated Test Battery. We used generalized additive models to examine nonlinear interaction effects of ELS severity and age on these outcomes. RESULTS Analyses revealed significant nonlinear interaction effects of ELS severity and age on NfL and GFAP serum concentrations, total and subcortical gray matter volume loss, increased third ventricular volume, and cognitive impairment. INTERPRETATION These findings suggest that ELS profoundly exacerbates peripheral, neurostructural, and cognitive markers of brain aging. Our results are critical for the development of novel early prevention strategies that target the impact of developmental stress on the brain to mitigate aging-related neurological diseases. ANN NEUROL 2025;97:642-656.
Collapse
Affiliation(s)
- Lara Fleck
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Claudia Buss
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
- Development, Health, and Disease Research ProgramUniversity of California, IrvineOrangeCA
- German Center for Mental HealthBerlinGermany
- German Center for Child and Adolescent HealthBerlinGermany
| | - Martin Bauer
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinExperimental and Clinical Research CenterBerlinGermany
| | - Maike Stein
- Department of Neurology with Experimental NeurologyCharité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Department of Neurology, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA
- Berlin Institute of Health at Charité—Universitätsmedizin BerlinDigital Health CenterBerlinGermany
- Center for Stroke Research BerlinCharité—Universitätsmedizin BerlinBerlinGermany
| | - Ralf Mekle
- Center for Stroke Research BerlinCharité—Universitätsmedizin BerlinBerlinGermany
| | - Lena Kock
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Heiko Klawitter
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Malvika Godara
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Judith Ramler
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
| | - Sonja Entringer
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
- Development, Health, and Disease Research ProgramUniversity of California, IrvineOrangeCA
- German Center for Mental HealthBerlinGermany
- German Center for Child and Adolescent HealthBerlinGermany
| | - Matthias Endres
- German Center for Mental HealthBerlinGermany
- Department of Neurology with Experimental NeurologyCharité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Center for Stroke Research BerlinCharité—Universitätsmedizin BerlinBerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, NeuroCure Cluster of ExcellenceBerlinGermany
- German Center for Neurodegenerative DiseasesBerlinGermany
- German Centre for Cardiovascular ResearchBerlinGermany
| | - Christine Heim
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Medical PsychologyBerlinGermany
- German Center for Mental HealthBerlinGermany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, NeuroCure Cluster of ExcellenceBerlinGermany
| |
Collapse
|
2
|
Coors A, Boenniger MM, Santos MLS, Lohner V, Koch A, Ettinger U, Aziz NA, Breteler MMB. Associations of Plasma Neurofilament Light Levels With Brain Microstructure and Macrostructure and Cognition in the Community-Based Rhineland Study. Neurology 2025; 104:e210278. [PMID: 39977717 DOI: 10.1212/wnl.0000000000210278] [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: 08/07/2024] [Accepted: 11/19/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Plasma neurofilament light chain (NfL) level is a sensitive yet aspecific marker of neurodegeneration. Its neuroanatomical and functional correlates in the general population are not fully elucidated. We thus assessed how brain's macrostructures and microstructures and cognitive function are related to plasma NfL levels in cognitively unimpaired adults over a wide age range. METHODS Our analyses were based on cross-sectional data from the Rhineland Study, a community-based prospective cohort study. This study includes people from the age of 30 onwards who live in 2 geographically defined areas in Bonn, Germany, and have sufficient command of the German language. Plasma NfL levels were measured using the Simoa platform and then log-transformed and adjusted for plate position, batch number, and Analyzer (HD-1 or HD-X). Brain imaging data were collected on a 3 Tesla scanner and included volumetric measures, metrics of the diffusion tensor and the neurite orientation dispersion and density imaging model, and white matter hyperintensity load. Memory performance, processing speed, and executive function were assessed using traditional cognitive tasks and an eye movement battery. We used multivariable regression models to assess the relations between brain structure and plasma NfL levels and between plasma NfL levels and cognitive performance. RESULTS The study sample consisted of 5,589 participants aged 30-95 years (mean age 55 ± 13.7 years, 56.1% women) without neurodegenerative diseases. Higher plasma NfL levels were associated with lower isotropic volume fraction (-0.030; 95% CI -0.051 to -0.010; pFDR = 0.011), lower neurite density index (ß = -0.031; 95% CI -0.053 to -0.008; pFDR = 0.014), and higher axial diffusivity (ß = 0.037; 95% CI 0.013-0.062; p = 0.005; pFDR = 0.011). The strongest association was with the orientation dispersion index (ß = -0.063; 95% CI -0.085 to -0.041; pFDR < 0.001). Furthermore, higher plasma NfL levels tended to be associated with a lower processing speed domain score (ß = -0.046; 95% CI -0.084 to -0.009; p = 0.014; pFDR = 0.056) and longer prosaccade latency (ß = 0.039; 95% CI 0.000-0.078; p = 0.049; pFDR = 0.480). DISCUSSION Higher plasma NfL levels mainly reflect worse white matter microstructural integrity, especially lower axonal density, in a relatively healthy, community-based sample. This suggests that plasma NfL levels allow for early detection of subtle differences in brain microstructure.
Collapse
Affiliation(s)
- Annabell Coors
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Cognitive Neuroscience Division, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Meta Miriam Boenniger
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department I for Internal Medicine, University of Cologne and University Hospital Cologne, Germany
| | - Marina L S Santos
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Valerie Lohner
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Alexandra Koch
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, Germany; and
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany
| |
Collapse
|
3
|
Rajput M, Malik IA, Methi A, Cortés Silva JA, Fey D, Wirths O, Fischer A, Wilting J, von Arnim CAF. Cognitive decline and neuroinflammation in a mouse model of obesity: An accelerating role of ageing. Brain Behav Immun 2025; 125:226-239. [PMID: 39730092 DOI: 10.1016/j.bbi.2024.12.154] [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: 06/14/2024] [Revised: 11/30/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024] Open
Abstract
Obesity, a pandemic, worldwide afflicts almost one billion people. Obesity and ageing share several pathological pathways leading to neurological disorders. However, due to a lack of suitable animal models, the long-term effects of obesity on age-related disorders- cognitive impairment and dementia have not yet been thoroughly investigated. Therefore, the current investigation focuses on developing a suitable model to explore the effects of obese-ageing. It also aims to determine whether obesity affects cognitive abilities in an age-dependent manner, and to identify a potential biomarker(s) for cognitive decline. Cognitive tests were carried out on 6-months and 1-year-old melanocortin-4 receptor (Mc4r)-deficient-obese and lean (wildtype) mice. Additionally, brains and sera were harvested for molecular, histological and serological analyses from 6, 12, and 24-months-old mice. Finally, RT-PCR was carried out after hippocampal mRNA sequencing. The cognitive tests revealed that 1-year-old obese mice have cognitive impairment along with underlying neurodegenerative changes, such as enlarged lateral ventricles. Serum neurofilament light chain (sNfL) levels were also elevated. Lipid accumulation and neuroinflammation were apparent besides, a compromised blood-brain barrier (BBB) indicated by altered junction protein gene expression. Differentially-expressed genes associated with cognitive decline were identified by mRNA sequencing of hippocampi. One such gene, Secreted Phosphoprotein 1 (Spp1) had markedly increased expression in cognitively-impaired obese mice. Our findings present an obese-aged mouse model of cognitive decline with neuroinflammation, reduced BBB-integrity and predisposing neurodegenerative changes. Obese-ageing accelerates the progression of cognitive impairment. Furthermore, Spp1 appears to be a potential biomarker for early diagnosis of neuropathological disorders.
Collapse
Affiliation(s)
- Mansi Rajput
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany.
| | - Ihtzaz Ahmed Malik
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany.
| | - Aditi Methi
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany.
| | - Jonathan Alexis Cortés Silva
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany.
| | - Dorothea Fey
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany.
| | - Oliver Wirths
- Department of Psychiatry, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075 Goettingen, Germany.
| | - André Fischer
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany; Department of Psychiatry, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075 Goettingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany.
| | - Jörg Wilting
- Institute of Anatomy and Embryology, University Medical Center Goettingen, Kreuzbergring 36, D-37075 Goettingen, Germany.
| | - Christine A F von Arnim
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany.
| |
Collapse
|
4
|
Kang Y, Feng Z, Zhang Q, Liu M, Li Y, Yang H, Zheng L, Cheng C, Zhou W, Lou D, Li X, Chen L, Feng Y, Duan X, Duan J, Yu M, Yang S, Liu Y, Wang X, Deng B, Liu C, Yao X, Zhu C, Liang C, Zeng X, Ren S, Li Q, Zhong Y, Yan Y, Meng H, Zhong Z, Zhang Y, Kang J, Luan X, Pan S, Wu Y, Li T, Song W, Zhang Y. Identification of circulating risk biomarkers for cognitive decline in a large community-based population in Chongqing China. Alzheimers Dement 2025; 21:e14443. [PMID: 39713874 PMCID: PMC11848162 DOI: 10.1002/alz.14443] [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: 08/30/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION This study aims to investigate the relationship between blood-based pathologies and established risk factors for cognitive decline in the community-based population of Chongqing, a region with significant aging. METHODS A total of 26,554 residents aged 50 years and older were recruited. Multinomial logistic regression models were applied to assess the risk factors of cognition levels. Propensity score matching and linear mixed effects models were used to evaluate the relationship between key risk factors and the circulating biomarkers. RESULTS Shared and distinct risk factors for MCI and dementia were identified. Age, lower education, medical history of stroke, hypertension, and epilepsy influenced mild cognitive impairment (MCI) progression to dementia. Correlations between key risk factors and circulating neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), amyloid β protein (Aβ)40, and Aβ42/Aβ40 ratio suggest underlying mechanisms contributing to cognitive impairment. DISCUSSION The common and distinct risk factors across cognitive decline stages emphasize the need for tailored interventions. The correlations with blood biomarkers provide insights into potential management targets. HIGHLIGHTS From a large community-based cohort study on the residents in Chongqing, we have identified that mild cognitive impairment (MCI) and dementia share several common risk factors, including age, female gender, rural living, lower education levels, and a medical history of stroke. However, each condition also has its own unique risk factors. Several factors contribute to the progression of MCI into dementia including age, education levels, occupation, and a medical history of hypertension and epilepsy. We discover the correlations between the risk factors for dementia and blood biomarkers that indicate the presence of axonal damage, glial activation, and Aβ pathology.
Collapse
|
5
|
Janus A, Dumas D, Le Douce J, Marie S, Pasculli G, Bambury P, Lemarchant S, Kremer P, Godfrin Y. Safety, Tolerability and Pharmacokinetic-Pharmacodynamic Relationship of NX210c Peptide in Healthy Elderly Volunteers: Randomized, Placebo-Controlled, Double-Blind, Multiple Ascending Dose Study. Neurol Ther 2025; 14:357-377. [PMID: 39708220 PMCID: PMC11762061 DOI: 10.1007/s40120-024-00691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Blood-brain barrier (BBB) integrity is fundamental to brain homeostasis, enabling control of substance exchange and safeguarding neurons against harmful toxins, pathogens, and immune cells that lead to dysregulation and inflammation involved in ageing and neurodegenerative diseases (NDD). The cyclized peptide NX210c is a thrombospondin type 1 repeat analogue derived from subcommissural organ-spondin. It exerts beneficial effects in animal models of NDD owing to its effects on neurons and endothelial cells. NX210c demonstrated a good safety profile in a single ascending dose phase 1a clinical study. The present multiple ascending dose phase 1b study was performed to evaluate the tolerability and pharmacological effects of repeated doses of NX210c in healthy elderly (age: > 55 years) volunteers. METHODS This was a randomized, placebo-controlled, double-blind study (EudraCT No. 2022-002868-76), investigating safety/tolerability, pharmacokinetics, and pharmacodynamics (including blood and cerebrospinal fluid biomarkers). Participants received 5 or 10 mg/kg NX210c or placebo (10-min infusion) thrice weekly for 4 weeks in an ascending dose fashion. Follow-up was conducted 2 weeks after last dosing. RESULTS The investigation included 29 participants. No serious adverse events were recorded and all adverse events were mild. Dedicated central nervous system testing did not reveal neurotoxicity. Biomarker evaluation showed a statistically significant reduction in blood claudin-5 and a trend toward reduction of blood homocysteine. In silico data modelling revealed salient pharmacokinetic-pharmacodynamic relationships, including reduction of claudin-5, neurofilament light chain, and SPARC-like protein 1 release, and degradation of homocysteine. CONCLUSION Multiple doses of NX210c exhibited a good safety profile, showed non-cumulative pharmacokinetics, and exerted pharmacodynamic effects on biomarkers linked to BBB integrity. The effects of NX210c on claudin-5 and biomarkers influencing BBB integrity-and the overarching brain protection it offers-provide a novel therapeutic strategy targeting an underlying driver of neurodegenerative conditions for which disease-modifying treatments are limited or not available.
Collapse
Affiliation(s)
- Annette Janus
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France.
| | - Daniël Dumas
- Centre for Human Drug Research (CHDR), Leiden, The Netherlands
- Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Juliette Le Douce
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | - Sébastien Marie
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | | | | | - Sighild Lemarchant
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
| | - Philip Kremer
- Centre for Human Drug Research (CHDR), Leiden, The Netherlands
- Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Yann Godfrin
- Axoltis Pharma, Bioparc Laennec, 60 Avenue Rockefeller, 69008, Lyon, France
- Godfrin Life-Sciences, Caluire-Et-Cuire, France
| |
Collapse
|
6
|
Lohner V, Perna L, Schöttker B, Perneczky R, Brenner H, Mons U. Associations of blood-based biomarkers of neurodegenerative diseases with mortality, cardio- and cerebrovascular events in persons with chronic coronary syndrome. Exp Gerontol 2025; 200:112684. [PMID: 39824235 DOI: 10.1016/j.exger.2025.112684] [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: 10/11/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
BACKGROUND In light of growing evidence highlighting interactions between cardiac and brain health, we investigated associations of biomarkers of neurodegenerative diseases with adverse outcomes (all-cause and cardiovascular mortality, major cardiovascular events, and stroke) in persons with chronic coronary syndrome (CCS). METHODS We used data from a cohort of persons with CCS for whom major adverse events were recorded over a follow-up of 20 years. We measured biomarkers of neurodegenerative diseases in baseline blood samples, using the Single-Molecule Array Technology on a HD-1 Analyzer. These include biomarkers of neuronal (neurofilament light chain (NfL) (n = 379)) and glial neurodegeneration (glial fibrillary acidic protein (GFAP) (n = 379)), and Alzheimer's disease pathology (phosphorylated tau181 (n = 379), total tau (n = 377), and amyloid β (Aβ40, Aβ42, Aβ42/Aβ40) (n = 377)). We applied Cox-proportional hazards models to evaluate associations of these biomarkers with adverse outcomes, adjusting for covariates and exploring interactions with apolipoprotein E (ApoE) ε4 genotype. RESULTS Participants with higher NfL levels had increased rates of all-cause and cardiovascular mortality (Hazard ratio per increase by one standard deviation (95 % confidence interval): all-cause mortality: 1.36 (1.10-1.68); cardiovascular mortality: 1.42 (1.05-1.93)). The Aβ40/Aβ42-ratio was linked to incident stroke (0.72 (0.52-1.00)). Associations of GFAP with all-cause mortality and incident stroke were depending on ApoE ε4 genotype. The other biomarkers were not significantly associated with the studied outcomes. CONCLUSIONS In persons with CSS, NfL and the Aβ40/Aβ42-ratio were related to mortality and incident stroke, respectively, whereas associations of GFAP with adverse outcomes varied by ApoE genotype. These biomarkers might play a role in linking aging, cardiovascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Valerie Lohner
- Cardiovascular Epidemiology of Aging, Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany.
| | - Laura Perna
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany; Division of Mental Health of Older Adults, Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; Network Aging Research, Heidelberg University, Heidelberg, Germany
| | - Robert Perneczky
- Division of Mental Health of Older Adults, Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Germany; Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Sheffield Institute for Translational Neurology (SITraN), University of Sheffield, Sheffield, UK
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; Network Aging Research, Heidelberg University, Heidelberg, Germany
| | - Ute Mons
- Cardiovascular Epidemiology of Aging, Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
7
|
Goeminne LJE, Vladimirova A, Eames A, Tyshkovskiy A, Argentieri MA, Ying K, Moqri M, Gladyshev VN. Plasma protein-based organ-specific aging and mortality models unveil diseases as accelerated aging of organismal systems. Cell Metab 2025; 37:205-222.e6. [PMID: 39488213 DOI: 10.1016/j.cmet.2024.10.005] [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: 04/30/2024] [Revised: 08/04/2024] [Accepted: 10/04/2024] [Indexed: 11/04/2024]
Abstract
Aging is a complex process manifesting at molecular, cellular, organ, and organismal levels. It leads to functional decline, disease, and ultimately death, but the relationship between these fundamental biomedical features remains elusive. By applying elastic net regularization to plasma proteome data of over 50,000 human subjects in the UK Biobank and other cohorts, we report interpretable organ-specific and conventional aging models trained on chronological age, mortality, and longitudinal proteome data. These models predict organ/system-specific disease and indicate that men age faster than women in most organs. Accelerated organ aging leads to diseases in these organs, and specific diets, lifestyles, professions, and medications influence organ aging rates. We then identify proteins driving these associations with organ-specific aging. Our analyses reveal that age-related chronic diseases epitomize accelerated organ- and system-specific aging, modifiable through environmental factors, advocating for both universal whole-organism and personalized organ/system-specific anti-aging interventions.
Collapse
Affiliation(s)
- Ludger J E Goeminne
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anastasiya Vladimirova
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alec Eames
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - M Austin Argentieri
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kejun Ying
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mahdi Moqri
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
8
|
Daniels N, Bindoff AD, Vickers JC, King AE, Collins JM. Vulnerability of neurofilament-expressing neurons in frontotemporal dementia. Mol Cell Neurosci 2024; 131:103974. [PMID: 39369804 DOI: 10.1016/j.mcn.2024.103974] [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: 06/19/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
Frontotemporal dementia (FTD) is an umbrella term for several early onset dementias, that are caused by frontotemporal lobar degeneration (FTLD), which involves the atrophy of the frontal and temporal lobes of the brain. Neuron loss in the frontal and temporal lobes is a characteristic feature of FTLD, however the selective vulnerability of different neuronal populations in this group of diseases is not fully understood. Neurofilament-expressing neurons have been shown to be selectively vulnerable in other neurodegenerative diseases, including Alzheimer's disease and amyotrophic lateral sclerosis, therefore we sought to investigate whether this neuronal population is vulnerable in FTLD. We also examined whether neuronal sub-type vulnerability differed between FTLD with TDP-43 inclusions (FTLD-TDP) and FTLD with tau inclusions (FTLD-Tau). Post-mortem human tissue from the superior frontal gyrus (SFG) of FTLD-TDP (n = 15), FTLD-Tau (n = 8) and aged Control cases (n = 6) was immunolabelled using antibodies against non-phosphorylated neurofilaments (SMI32 antibody), calretinin and NeuN, to explore neuronal cell loss. The presence of non-phosphorylated neurofilament immunolabelling in axons of the SFG white matter was also quantified as a measure of axon pathology, as axonal neurofilaments are normally phosphorylated. We demonstrate the selective loss of neurofilament-expressing neurons in both FTLD-TDP and FTLD-Tau cases compared to aged Controls. We also show that non-phosphorylated neurofilament axonal pathology in the SFG white matter was associated with increasing age, but not FTLD. This data suggests neurofilament-expressing neurons are vulnerable in both FTLD-TDP and FTLD-Tau.
Collapse
Affiliation(s)
- Nina Daniels
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia.
| | - Aidan D Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Jessica M Collins
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
9
|
Liu X, Chen X, Chen J. Relationship between serum neurofilament light chain protein and depression: A nationwide survey and Mendelian randomization study. J Affect Disord 2024; 366:162-171. [PMID: 39197554 DOI: 10.1016/j.jad.2024.08.130] [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: 05/27/2024] [Revised: 07/22/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Investigating the link between serum neurofilament protein (sNfL) levels and depression remains an area of limited understanding. This study explores the correlation in US adults and employs Mendelian randomization (MR) to ascertain causality. METHODS Our cross-sectional study analyzed data from participants aged 20 and above in the National Health and Nutrition Examination Survey (2013-2014). We employed a weighted multiple logistic regression model to examine the relationship between ln (sNfL) and depression. Restricted cubic splines (RCS) were used to visualize non-linear relationships. Stratified analyses examined associations between ln(sNfL) and depression in different subgroups. Subsequently, we conducted a two-sample bidirectional Mendelian randomization (MR) to assess the causal relationship between sNfL and depression. The inverse variance-weighted (IVW) method was utilized as the primary analysis. RESULTS Among 1765 participants (mean age 45.19 years; 49.37 % male), 166 had depression with a Patient Health Questionnaire (PHQ-9) score ≥ 10. After adjusting for covariates, a positive correlation remained between sNfL and depression (OR 1.511, 95 % CI: 1.050-2.175). RCS curves indicated a non-linear association, with a turning point at 2.76 pg/ml. Stratified analyses revealed positive correlations in specific subgroups, with interactions involving age, race, family income, recreational activity, and ln(sNfL). MR using IVW found no significant causal relationship between sNfL and depression genetically (OR = 0.956, 95 % CI: 0.878-1.042), with reverse analysis yielding similar results (OR = 0.897, 95 % CI: 0.756-1.065). CONCLUSIONS This cross-sectional study highlights a significant correlation between ln(sNfL) and depression. However, MR results indicate no causal relationship between sNfL and depression.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, China.
| | - Xiong Chen
- Department of Mental Health Centre, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Jun Chen
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
10
|
Hofmann A, Häsler LM, Lambert M, Kaeser SA, Gräber-Sultan S, Obermüller U, Kuder-Buletta E, la Fougere C, Laske C, Vöglein J, Levin J, Fox NC, Ryan NS, Zetterberg H, Llibre-Guerra JJ, Perrin RJ, Ibanez L, Schofield PR, Brooks WS, Day GS, Farlow MR, Allegri RF, Chrem Mendez P, Ikeuchi T, Kasuga K, Lee JH, Roh JH, Mori H, Lopera F, Bateman RJ, McDade E, Gordon BA, Chhatwal JP, Jucker M, Schultz SA. Comparative neurofilament light chain trajectories in CSF and plasma in autosomal dominant Alzheimer's disease. Nat Commun 2024; 15:9982. [PMID: 39557867 PMCID: PMC11574007 DOI: 10.1038/s41467-024-52937-8] [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: 04/11/2024] [Accepted: 09/25/2024] [Indexed: 11/20/2024] Open
Abstract
Disease-modifying therapies for Alzheimer's disease (AD) are likely to be most beneficial when initiated in the presymptomatic phase. To track the benefit of such interventions, fluid biomarkers are of great importance, with neurofilament light chain protein (NfL) showing promise for monitoring neurodegeneration and predicting cognitive outcomes. Here, we update and complement previous findings from the Dominantly Inherited Alzheimer Network Observational Study by using matched cross-sectional and longitudinal cerebrospinal fluid (CSF) and plasma samples from 567 individuals, allowing timely comparative analyses of CSF and blood trajectories across the entire disease spectrum. CSF and plasma trajectories were similar at presymptomatic stages, discriminating mutation carriers from non-carrier controls 10-20 years before the estimated onset of clinical symptoms, depending on the statistical model used. However, after symptom onset the rate of change in CSF NfL continued to increase steadily, whereas the rate of change in plasma NfL leveled off. Both plasma and CSF NfL changes were associated with grey-matter atrophy, but not with Aβ-PET changes, supporting a temporal decoupling of Aβ deposition and neurodegeneration. These observations support NfL in both CSF and blood as an early marker of neurodegeneration but suggest that NfL measured in the CSF may be better suited for monitoring clinical trial outcomes in symptomatic AD patients.
Collapse
Affiliation(s)
- Anna Hofmann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lisa M Häsler
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Marius Lambert
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Stephan A Kaeser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | - Ulrike Obermüller
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | - Christian la Fougere
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig Maximilians-Universität München, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig Maximilians-Universität München, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Henrik Zetterberg
- Department 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
| | - Jorge J Llibre-Guerra
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard J Perrin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura Ibanez
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter R Schofield
- Neuroscience Research Australia, Randwick, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - William S Brooks
- Neuroscience Research Australia, Randwick, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health Sydney, University of New South Wales, Sydney, Australia
| | - Gregory S Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Martin R Farlow
- Indiana Alzheimer Disease Center and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Takeshi Ikeuchi
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Kensaku Kasuga
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jee Hoon Roh
- Departments of Neurology and Physiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Hiroshi Mori
- Faculty of Medicine, Osaka Metropolitan University, Nagaoka Sutoku University, Osaka, Japan
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia (GNA), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
- Brigham and Women's Hospital Boston, Boston, MA, USA
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.
| | - Stephanie A Schultz
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
11
|
Cai Y, Wei Q, Wang S, Lu X. All-Cause Mortality Differentials by Diabetes Status and Serum Neurofilament Light-Chain Levels in US General Adults. J Clin Endocrinol Metab 2024; 109:e2246-e2254. [PMID: 38412312 DOI: 10.1210/clinem/dgae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 02/29/2024]
Abstract
CONTEXT Neurofilament light chains (sNFLs) increase in patients with diabetes (DM) and are associated with death. OBJECTIVE This work aimed to examine whether sNFL mediates associations of DM with all-cause mortality and the extent of interaction or joint relations of sNFL and DM with mortality. METHODS This population-based cohort study was conducted using the 2013 to 2014 cycle of the National Health and Nutrition Examination Survey. A total of 2071 adults aged 20 to 75 years with sNFL measurements were included. sNFL was lg-transformed (LgNfl). Participants were included whose LgNfl was higher than 1.48 pg/mL or who were diagnosed with DM. All-cause mortality was the primary outcome obtained through linkage to registries. RESULTS During a median follow-up of 6.1years, 85 participants died. Incidence rates (per 1000 person-years [95% CI]) of all-cause mortality were 27.78 (19.98∼35.58) in adults with LgNfl greater than 1.48 pg/mL and DM, 9.01 (1.99∼16.03) in adults with LgNfl greater than 1.48 pg/mL but no DM, 3.07 (1.01∼5.13) in adults with DM and LgNfl less than or equal to 1.48 pg/mL, and 2.21 (1.15∼3.27) in adults without DM and LgNfl less than or equal to 1.48 pg/mL. Significant interaction but not mediation was observed between LgNfl and DM. Compared with adults without DM and LgNfl less than or equal to 1.48 pg/mL, those with DM and LgNfl greater than 1.48 pg/mL had higher risks of all-cause mortality (hazard ratio; 95% CI, 7.06; 3.52∼14.16). CONCLUSION In general US adults with DM, elevated sNFLs were associated with higher all-cause mortality specifically, supporting an important role of sNFLs in predicting health outcome in individuals with DM.
Collapse
Affiliation(s)
- Yingyuan Cai
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Qian Wei
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Shan Wang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xiaowei Lu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| |
Collapse
|
12
|
Holmegaard L, Jensen C, Pedersen A, Blomstrand C, Blennow K, Zetterberg H, Jood K, Jern C. Circulating levels of neurofilament light chain as a biomarker of infarct and white matter hyperintensity volumes after ischemic stroke. Sci Rep 2024; 14:16180. [PMID: 39003344 PMCID: PMC11246414 DOI: 10.1038/s41598-024-67232-1] [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: 01/11/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024] Open
Abstract
Serum neurofilament light chain protein (sNfL) shows promise as a biomarker for infarct size in acute ischemic stroke and for monitoring cerebral small vessel disease (cSVD). However, distinguishing the cSVD contribution after stroke may not be possible due to post-stroke sNfL increase. Additionally, it remains unclear if etiologic subtype differences exist. We measured infarct and white matter hyperintensity (WMH) volumes using MRI at the index stroke in ischemic stroke patients (n = 316, mean age 53 years, 65% males) and at 7-year follow-up (n = 187). Serum NfL concentration was measured in the acute phase (n = 235), at 3-months (n = 288), and 7-years (n = 190) post stroke. In multivariable regression, acute and 3-month sNfL concentrations were associated with infarct volume and time since stroke, but not with stroke etiology or infarct location. Seven years post-stroke, sNfL was associated with WMHs and age, but not with stroke etiology. Nonlinear regression estimated that sNfL peaks around 1 month, and declines by 50% at 3 months, and 99% at 9 months. We conclude that sNfL can indicate infarct volume and time since brain injury in the acute and subacute phases after stroke. Due to the significant post-stroke sNfL increase, several months are needed for reliable assessment of cSVD activity.
Collapse
Affiliation(s)
- Lukas Holmegaard
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Neurology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Christer Jensen
- Department of Radiology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Radiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Annie Pedersen
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Christian Blomstrand
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Region Västra Götaland, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Region Västra Götaland, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Katarina Jood
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neurology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Christina Jern
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
13
|
Beydoun MA, Noren Hooten N, Georgescu MF, Beydoun HA, Eid SM, Fanelli-Kuczmarski MT, Evans MK, Zonderman AB. Serum neurofilament light chain as a prognostic marker of all-cause mortality in a national sample of US adults. Eur J Epidemiol 2024; 39:795-809. [PMID: 38771439 PMCID: PMC11343803 DOI: 10.1007/s10654-024-01131-7] [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: 03/04/2023] [Accepted: 05/01/2024] [Indexed: 05/22/2024]
Abstract
Neurofilament light chain (NfL) is a neuron-specific structural protein released into the extracellular space, including body fluids, upon neuroaxonal damage. Despite evidence of a link in neurological disorders, few studies have examined the association of serum NfL with mortality in population-based studies. Data from the National Health and Nutrition Survey were utilized including 2,071 Non-Hispanic White, Non-Hispanic Black and Hispanic adult participants and adult participants of other ethnic groups (20-85 years) with serum NfL measurements who were followed for ≤ 6 years till 2019. We tested the association of serum NfL with mortality in the overall population and stratified by sex with the addition of potential interactive and mediating effects of cardio-metabolic risk factors and nutritional biomarkers. Elevated serum NfL levels (above median group) were associated with mortality risk compared to the below median NfL group in the overall sample (P = 0.010), with trends observed within each sex group (P < 0.10). When examining Loge NfL as a continuum, one standard deviation of Loge NfL was associated with an increased mortality risk (HR = 1.88, 95% CI 1.60-2.20, P < 0.001) in the reduced model adjusted for age, sex, race, and poverty income ratio; a finding only slightly attenuated with the adjustment of lifestyle and health-related factors. Four-way decomposition indicated that there was, among others, mediated interaction between NfL and HbA1c and a pure inconsistent mediation with 25(OH)D3 in predicting all-cause mortality, in models adjusted for all other covariates. Furthermore, urinary albumin-to-creatinine ratio interacted synergistically with NfL in relation to mortality risk both on the additive and multiplicative scales. These data indicate that elevated serum NfL levels were associated with all-cause mortality in a nationally representative sample of US adults.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA.
- NIH Biomedical Research Center, National Institute on Aging, IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA.
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Hind A Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA, USA
| | - Shaker M Eid
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| |
Collapse
|
14
|
Perneczky R, Hansen N, Hofmann A, Laske C, Priller J, Grimmer T, Frölich L, Düzel E, Jessen F, Wiltfang J. Blood-Based Biomarkers for Early Alzheimer's Disease Diagnosis in Real-World Settings. Methods Mol Biol 2024; 2785:3-14. [PMID: 38427184 DOI: 10.1007/978-1-0716-3774-6_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] [Indexed: 03/02/2024]
Abstract
As our knowledge about the biology of Alzheimer's disease (AD) expands and we recognize the significance of early intervention for effective treatment, there is a shift in focus toward detecting the disease at an early stage. AD is characterized by the accumulation of misfolded amyloid-β (Aβ) and phosphorylated tau proteins in the brain, leading to the formation of senile plaques and neurofibrillary tangles. While a definitive diagnosis of AD can only be confirmed through autopsy by examining these pathological features, there are now reliable methods available for diagnosing the disease in living individuals. These methods involve analyzing cerebrospinal fluid and using positron emission tomography to accurately assess the presence of Aβ and tau proteins. While these diagnostic markers have shown high accuracy in memory-clinic populations, they do have limitations such as the requirement for invasive lumbar puncture or exposure to ionizing radiation. Additionally, they are not easily accessible outside of specialized healthcare settings. Blood-based biomarkers of the core pathological features of AD are being developed, showing promise for less invasive, scalable identification of AD cases in the community. The advantages for the healthcare systems of this development are obvious, but the diagnostic performance of blood-based biomarkers in broader, non-selected populations outside of retrospective analyses and research cohorts still requires further investigation, including the combination with more effective neuropsychological assessments such as digital cognitive test solutions.
Collapse
Affiliation(s)
- Robert Perneczky
- Department of Psychiatry and Psychotherapy, LMU Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK.
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK.
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Anna Hofmann
- Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
- Department of Neurology, University Hospital Tuebingen, Tuebingen, Germany
- German Center for Neurodegenerative Diseases (DZNE) Tuebingen, Tuebingen, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE) Tuebingen, Tuebingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tuebingen, Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charite University Medicine, Berlin, Germany
- Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Emrah Düzel
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Magdeburg, Germany
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE) Bonn, Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE) Goettingen, Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
15
|
Liu X, Chen J, Meng C, Zhou L, Liu Y. Serum neurofilament light chain and cognition decline in US elderly: A cross-sectional study. Ann Clin Transl Neurol 2024; 11:17-29. [PMID: 37902309 PMCID: PMC10791034 DOI: 10.1002/acn3.51929] [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: 07/14/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023] Open
Abstract
OBJECTIVE Early identification of cognitive impairment in neurodegenerative diseases like Alzheimer's disease (AD) is crucial. Neurofilament, a potential biomarker for neurological disorders, has gained attention. Our study aims to investigate the relationship between serum neurofilament light (sNfL) levels and cognitive function in elderly individuals in the United States. METHODS This cross-sectional study analyzed data from participants aged 60 and above in the National Health and Nutrition Examination Survey (2013-2014). We collected sNfL levels, cognitive function tests, sociodemographic characteristics, comorbidities, and other variables. Weighted multiple linear regression models examined the relationship between ln(sNfL) and cognitive scores. Restricted cubic spline (RCS) visualization explored nonlinear relationships. The stratified analysis examined subgroups' ln(sNfL) and cognitive function association. RESULTS The study included 446 participants (47.73% male). Participants with ln(sNfL) levels between 2.58 and 2.81 pg/mL (second quintile) performed relatively well in cognitive tests. After adjusting for multiple factors, ln(sNfL) levels were negatively correlated with cognitive function, with adjusted β (95% CI) as follows: immediate recall test (IRT): -0.763 (-1.301 to -0.224), delayed recall test (DRT): -0.308 (-0.576 to -0.04), animal fluency test (AFT): -1.616 (-2.639 to -0.594), and digit symbol substitution test (DSST): -2.790 (-4.369 to -1.21). RCS curves showed nonlinear relationships between ln(sNfL) and DRT, AFT, with inflection points around 2.7 pg/mL. The stratified analysis revealed a negative correlation between ln(sNfL) and cognition in specific subgroups with distinct features, with an interaction between diabetes and ln(sNfL). INTERPRETATION Higher sNfL levels are associated with poorer cognitive function in the elderly population of the United States. sNfL shows promise as a potential biomarker for early identification of cognitive decline.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Jun Chen
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Chen Meng
- Department of Anesthesiology, Taihe HospitalHubei University of MedicineShiyanHubeiChina
| | - Lan Zhou
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Yong Liu
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| |
Collapse
|
16
|
Chen X, Lin Y, Wei K. Elevated Serum Neurofilament Light Chain Levels Are Associated With All-Cause Mortality: Evidence From National Health and Nutrition Examination Survey. J Gerontol A Biol Sci Med Sci 2023; 78:2382-2386. [PMID: 36811342 DOI: 10.1093/gerona/glad058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Several studies have reported the association between blood neurofilament light chain (NfL) levels and all-cause mortality. However, the generalizability of these findings in general adults remains unclear. The study aimed to examine the association between serum NfL and all-cause mortality in a nationally representative population. METHODS Longitudinal data were obtained from 2 071 participants aged 20-75 years in the National Health and Nutrition Examination Survey 2013-14 cycle. Serum NfL levels were measured by using a novel, high-throughput acridinium-ester immunoassay. Kaplan-Meier curves, multivariate Cox regression analysis, and restricted cubic spline regression were employed to investigate the association between serum NfL and all-cause mortality. RESULTS Over a median follow-up of 73 (interquartile range = 12) months, 85 (3.50%) participants died. After adjustment for sociodemographic characteristics, lifestyle variables, comorbidity, body mass index, and estimated glomerular filtration rate, elevated serum NfL levels were still significantly associated with a higher risk of all-cause mortality (hazard ratio = 2.45, 95% confidence interval = 1.89-3.18 for per lnNfL increase) in a linear manner. CONCLUSIONS Our findings suggest that circulating levels of NfL may serve as a biomarker of mortality risk in a nationally representative population.
Collapse
Affiliation(s)
- Xiaotong Chen
- Department of Laboratory Medicine, Jing'an District Central Hospital of Shanghai, Jing'an Branch Affiliated to Huashan Hospital, Fudan University, Shanghai, China
| | - Yong Lin
- Department of Laboratory Medicine, Jing'an District Central Hospital of Shanghai, Jing'an Branch Affiliated to Huashan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Kai Wei
- Department of Pharmacy, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
17
|
Hahn O, Foltz AG, Atkins M, Kedir B, Moran-Losada P, Guldner IH, Munson C, Kern F, Pálovics R, Lu N, Zhang H, Kaur A, Hull J, Huguenard JR, Grönke S, Lehallier B, Partridge L, Keller A, Wyss-Coray T. Atlas of the aging mouse brain reveals white matter as vulnerable foci. Cell 2023; 186:4117-4133.e22. [PMID: 37591239 PMCID: PMC10528304 DOI: 10.1016/j.cell.2023.07.027] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 05/17/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023]
Abstract
Aging is the key risk factor for cognitive decline, yet the molecular changes underlying brain aging remain poorly understood. Here, we conducted spatiotemporal RNA sequencing of the mouse brain, profiling 1,076 samples from 15 regions across 7 ages and 2 rejuvenation interventions. Our analysis identified a brain-wide gene signature of aging in glial cells, which exhibited spatially defined changes in magnitude. By integrating spatial and single-nucleus transcriptomics, we found that glial aging was particularly accelerated in white matter compared with cortical regions, whereas specialized neuronal populations showed region-specific expression changes. Rejuvenation interventions, including young plasma injection and dietary restriction, exhibited distinct effects on gene expression in specific brain regions. Furthermore, we discovered differential gene expression patterns associated with three human neurodegenerative diseases, highlighting the importance of regional aging as a potential modulator of disease. Our findings identify molecular foci of brain aging, providing a foundation to target age-related cognitive decline.
Collapse
Affiliation(s)
- Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Aulden G Foltz
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Micaiah Atkins
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Blen Kedir
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Patricia Moran-Losada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ian H Guldner
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Christy Munson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA; Vilcek Institute of Graduate Biomedical Sciences, NYU Langone Health, New York City, NY, USA
| | - Fabian Kern
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Róbert Pálovics
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Nannan Lu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Hui Zhang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Achint Kaur
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacob Hull
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Andreas Keller
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University, Stanford, CA, USA; Stanford University, The Phil and Penny Knight Initiative for Brain Resilience, Stanford, CA, USA.
| |
Collapse
|
18
|
Self WK, Holtzman DM. Emerging diagnostics and therapeutics for Alzheimer disease. Nat Med 2023; 29:2187-2199. [PMID: 37667136 DOI: 10.1038/s41591-023-02505-2] [Citation(s) in RCA: 136] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/18/2023] [Indexed: 09/06/2023]
Abstract
Alzheimer disease (AD) is the most common contributor to dementia in the world, but strategies that slow or prevent its clinical progression have largely remained elusive, until recently. This Review highlights the latest advances in biomarker technologies and therapeutic development to improve AD diagnosis and treatment. We review recent results that enable pathological staging of AD with neuroimaging and fluid-based biomarkers, with a particular emphasis on the role of amyloid, tau and neuroinflammation in disease pathogenesis. We discuss the lessons learned from randomized controlled trials, including some supporting the proposal that certain anti-amyloid antibodies slow cognitive decline during the mildly symptomatic phase of AD. In addition, we highlight evidence for newly identified therapeutic targets that may be able to modify AD pathogenesis and progression. Collectively, these recent discoveries-and the research directions that they open-have the potential to move AD clinical care toward disease-modifying treatment strategies with maximal benefits for patients.
Collapse
Affiliation(s)
- Wade K Self
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, 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, USA.
| |
Collapse
|
19
|
Jones KT, Gallen CL, Ostrand AE, Rojas JC, Wais P, Rini J, Chan B, Lago AL, Boxer A, Zhao M, Gazzaley A, Zanto TP. Gamma neuromodulation improves episodic memory and its associated network in amnestic mild cognitive impairment: a pilot study. Neurobiol Aging 2023; 129:72-88. [PMID: 37276822 PMCID: PMC10583532 DOI: 10.1016/j.neurobiolaging.2023.04.005] [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: 09/06/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 06/07/2023]
Abstract
Amnestic mild cognitive impairment (aMCI) is a predementia stage of Alzheimer's disease associated with dysfunctional episodic memory and limited treatment options. We aimed to characterize feasibility, clinical, and biomarker effects of noninvasive neurostimulation for aMCI. 13 individuals with aMCI received eight 60-minute sessions of 40-Hz (gamma) transcranial alternating current stimulation (tACS) targeting regions related to episodic memory processing. Feasibility, episodic memory, and plasma Alzheimer's disease biomarkers were assessed. Neuroplastic changes were characterized by resting-state functional connectivity (RSFC) and neuronal excitatory/inhibitory balance. Gamma tACS was feasible and aMCI participants demonstrated improvement in multiple metrics of episodic memory, but no changes in biomarkers. Improvements in episodic memory were most pronounced in participants who had the highest modeled tACS-induced electric fields and exhibited the greatest changes in RSFC. Increased RSFC was also associated with greater hippocampal excitability and higher baseline white matter integrity. This study highlights initial feasibility and the potential of gamma tACS to rescue episodic memory in an aMCI population by modulating connectivity and excitability within an episodic memory network.
Collapse
Affiliation(s)
- Kevin T Jones
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA.
| | - Courtney L Gallen
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Avery E Ostrand
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Julio C Rojas
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Peter Wais
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - James Rini
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Brandon Chan
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Argentina Lario Lago
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Adam Boxer
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Min Zhao
- Departments of Ophthalmology and Vision Science and Dermatology, Institute for Regenerative Cures, University of California-Davis, Davis, CA
| | - Adam Gazzaley
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA; Departments of Physiology and Psychiatry, University of California-San Francisco, San Francisco, CA
| | - Theodore P Zanto
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA.
| |
Collapse
|
20
|
Li Y, Patel M, Baroudi J, Wu M, Gatti S, Liang M, Wipf P, Badawi Y, Meriney SD. A cross-sectional study of ageing at the mouse neuromuscular junction and effects of an experimental therapeutic approach for dynapenia. J Physiol 2023; 601:4135-4150. [PMID: 37606613 DOI: 10.1113/jp284749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 08/08/2023] [Indexed: 08/23/2023] Open
Abstract
Despite prior efforts to understand and target dynapenia (age-induced loss of muscle strength), this condition remains a major challenge that reduces the quality of life in the aged population. We have focused on the neuromuscular junction (NMJ) where changes in structure and function have rarely been systematically studied as a dynamic and progressive process. Our cross-sectional study found neurotransmission at the male mouse NMJ to be biphasic, displaying an early increase followed by a later decrease, and this phenotype was associated with structural changes to the NMJ. A cross-sectional characterization showed that age-induced alterations fell into four age groups: young adult (3-6 months), adult (7-18 months), early aged (19-24 months), and later aged (25-30 months). We then utilized a small molecule therapeutic candidate, GV-58, applied acutely during the later aged stage to combat age-induced reductions in transmitter release by increasing calcium influx during an action potential, which resulted in a significant increase in transmitter release. This comprehensive study of neuromuscular ageing at the NMJ will enable future research to target critical time points for therapeutic intervention. KEY POINTS: Age-induced frailty and falls are the leading causes of injury-related death and are caused by an age-induced loss of muscle strength due to a combination of neurological and muscular changes. A cross-sectional approach was used to study age-induced changes to the neuromuscular junction in a mouse model, and physiological changes that were biphasic over the ageing time course were found. Changes in physiology at the neuromuscular junction were correlated with alterations in neuromuscular junction morphology. An acutely applied positive allosteric gating modifier of presynaptic voltage-gated calcium channels was tested as a candidate therapeutic strategy that could increase transmitter release at aged neuromuscular junctions. These results provide a detailed time course of age-induced changes at the neuromuscular junction in a mouse model and test a candidate therapeutic strategy for weakness.
Collapse
Affiliation(s)
- Y Li
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Patel
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Baroudi
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Wu
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - S Gatti
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Liang
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - P Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Y Badawi
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen D Meriney
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Monzó C, Gkioni L, Beyer A, Valenzano DR, Grönke S, Partridge L. Dietary restriction mitigates the age-associated decline in mouse B cell receptor repertoire diversity. Cell Rep 2023; 42:112722. [PMID: 37384530 PMCID: PMC10391628 DOI: 10.1016/j.celrep.2023.112722] [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: 01/16/2023] [Revised: 05/07/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023] Open
Abstract
Aging impairs the capacity to respond to novel antigens, reducing immune protection against pathogens and vaccine efficacy. Dietary restriction (DR) extends life- and health span in diverse animals. However, little is known about the capacity of DR to combat the decline in immune function. Here, we study the changes in B cell receptor (BCR) repertoire during aging in DR and control mice. By sequencing the variable region of the BCR heavy chain in the spleen, we show that DR preserves diversity and attenuates the increase in clonal expansions throughout aging. Remarkably, mice starting DR in mid-life have repertoire diversity and clonal expansion rates indistinguishable from chronic DR mice. In contrast, in the intestine, these traits are unaffected by either age or DR. Reduced within-individual B cell repertoire diversity and increased clonal expansions are correlated with higher morbidity, suggesting a potential contribution of B cell repertoire dynamics to health during aging.
Collapse
Affiliation(s)
- Carolina Monzó
- Department Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, North Rhine Westphalia, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Age-Associated Diseases (CECAD), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50931 Cologne, Germany
| | - Lisonia Gkioni
- Department Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, North Rhine Westphalia, Germany
| | - Andreas Beyer
- Cologne Excellence Cluster on Cellular Stress Responses in Age-Associated Diseases (CECAD), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, 50931 Cologne, Germany
| | - Dario Riccardo Valenzano
- Microbiome-Host Interactions in Ageing Group, Max Planck Institute for Biology of Ageing, 50931 Cologne, North Rhine Westphalia, Germany; Evolutionary Biology/Microbiome-Host Interactions in Aging Group: Fritz Lipmann Institute - Leibniz Institute on Aging, 07745 Jena, Thuringia, Germany.
| | - Sebastian Grönke
- Department Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, North Rhine Westphalia, Germany.
| | - Linda Partridge
- Department Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, North Rhine Westphalia, Germany; Genetics, Evolution & Environment Group, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK.
| |
Collapse
|
22
|
Jia YJ, Wang J, Ren JR, Chan P, Chen S, Chen XC, Chhetri JK, Guo J, Guo Q, Jin L, Liu Q, Liu Q, Ma W, Mao Z, Song M, Song W, Tang Y, Wang D, Wang P, Xiong L, Ye K, Zhang J, Zhang W, Zhang X, Zhang Y, Zhang Z, Zhang Z, Zheng J, Liu GH, Eve Sun Y, Wang YJ, Pei G. A framework of biomarkers for brain aging: a consensus statement by the Aging Biomarker Consortium. LIFE MEDICINE 2023; 2:lnad017. [PMID: 39872296 PMCID: PMC11749242 DOI: 10.1093/lifemedi/lnad017] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/03/2023] [Indexed: 01/30/2025]
Abstract
China and the world are facing severe population aging and an increasing burden of age-related diseases. Aging of the brain causes major age-related brain diseases, such as neurodegenerative diseases and stroke. Identifying biomarkers for the effective assessment of brain aging and establishing a brain aging assessment system could facilitate the development of brain aging intervention strategies and the effective prevention and treatment of aging-related brain diseases. Thus, experts from the Aging Biomarker Consortium (ABC) have combined the latest research results and practical experience to recommend brain aging biomarkers and form an expert consensus, aiming to provide a basis for assessing the degree of brain aging and conducting brain-aging-related research with the ultimate goal of improving the brain health of elderly individuals in both China and the world.
Collapse
Affiliation(s)
| | - Yu-Juan Jia
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jun-Rong Ren
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Piu Chan
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Chun Chen
- Department of Neurology, Union Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Jagadish K Chhetri
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Junhong Guo
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lingjing Jin
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons’ Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University, School of Medicine, Shanghai 200092, China
| | - Qiang Liu
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Qiang Liu
- Department of Neurology, Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenlin Ma
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weihong Song
- Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Clinical Research Center for Mental Disorders, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Oujiang Laboratory, Wenzhou, Zhejiang 325035, China
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China
| | - Difei Wang
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Peijun Wang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, Tongji University, Shanghai 200434, China
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Xiaoqing Zhang
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Yunwu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Zhuohua Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jialin Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Key Laboratory of Ageing and Brain Disease, Chongqing 400042, China
| | - Gang Pei
- Collaborative Innovation Center for Brain Science, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| |
Collapse
|
23
|
Ciardullo S, Muraca E, Bianconi E, Ronchetti C, Cannistraci R, Rossi L, Perra S, Zerbini F, Perseghin G. Serum neurofilament light chain levels are associated with all-cause mortality in the general US population. J Neurol 2023:10.1007/s00415-023-11739-6. [PMID: 37085649 DOI: 10.1007/s00415-023-11739-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 04/23/2023]
Abstract
INTRODUCTION Serum neurofilament light chain (sNfL) levels are biomarkers of neuro-axonal injury in multiple neurological diseases. Little is known on their potential role as prognostic markers in people without known neurological conditions. OBJECTIVE The aim of this study is to evaluate the association between sNfL levels and all-cause mortality in a general population setting. METHODS sNfL levels were measured in 2071 people aged 25-75 years from the general US population that participated in the 2013-2014 cycles of the National Health and Nutrition Examination Survey (NHANES). Cognitive function was evaluated in a subset of participants aged 60-75 years using the Consortium to Establish a Registry for Alzheimer's Disease-Word Learning test, the Animal Fluency test and the Digit Symbol Substitution test. We applied Cox proportional hazard models adjusted for several potential confounders to evaluate the association between sNfL and all-cause mortality through December 2019 by linking NHANES data with data from the National Death Index. RESULTS In a cross-sectional analysis, higher sNfL levels were associated with worse performance in all three cognitive function tests. Over a median follow-up of 6.1 years, 85 participants died. In a multivariable model adjusted for age, sex, race-ethnicity, diabetes, chronic kidney disease, harmful alcohol consumption, cigarette smoke and prevalent cardiovascular disease, higher sNfL levels were significantly and positively associated with all-cause mortality (HR per unit increase in log-transformed sNfL: 2.46, 95% CI 1.77-3.43, p < 0.001). Results were robust when analyses were stratified according to age, sex, body mass index and kidney function. CONCLUSION We found a positive association between sNfL levels and mortality in the general US population. Further studies are needed to understand the biological mechanisms underlying this association.
Collapse
Affiliation(s)
- Stefano Ciardullo
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy.
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy.
| | - Emanuele Muraca
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Eleonora Bianconi
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Celeste Ronchetti
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Rosa Cannistraci
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Laura Rossi
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Silvia Perra
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Francesca Zerbini
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Gianluca Perseghin
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| |
Collapse
|
24
|
Cumplido-Mayoral I, García-Prat M, Operto G, Falcon C, Shekari M, Cacciaglia R, Milà-Alomà M, Lorenzini L, Ingala S, Meije Wink A, Mutsaerts HJMM, Minguillón C, Fauria K, Molinuevo JL, Haller S, Chetelat G, Waldman A, Schwarz AJ, Barkhof F, Suridjan I, Kollmorgen G, Bayfield A, Zetterberg H, Blennow K, Suárez-Calvet M, Vilaplana V, Gispert JD. Biological brain age prediction using machine learning on structural neuroimaging data: Multi-cohort validation against biomarkers of Alzheimer's disease and neurodegeneration stratified by sex. eLife 2023; 12:e81067. [PMID: 37067031 PMCID: PMC10181824 DOI: 10.7554/elife.81067] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 04/10/2023] [Indexed: 04/18/2023] Open
Abstract
Brain-age can be inferred from structural neuroimaging and compared to chronological age (brain-age delta) as a marker of biological brain aging. Accelerated aging has been found in neurodegenerative disorders like Alzheimer's disease (AD), but its validation against markers of neurodegeneration and AD is lacking. Here, imaging-derived measures from the UK Biobank dataset (N=22,661) were used to predict brain-age in 2,314 cognitively unimpaired (CU) individuals at higher risk of AD and mild cognitive impaired (MCI) patients from four independent cohorts with available biomarker data: ALFA+, ADNI, EPAD, and OASIS. Brain-age delta was associated with abnormal amyloid-β, more advanced stages (AT) of AD pathology and APOE-ε4 status. Brain-age delta was positively associated with plasma neurofilament light, a marker of neurodegeneration, and sex differences in the brain effects of this marker were found. These results validate brain-age delta as a non-invasive marker of biological brain aging in non-demented individuals with abnormal levels of biomarkers of AD and axonal injury.
Collapse
Affiliation(s)
- Irene Cumplido-Mayoral
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Marina García-Prat
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
| | - Grégory Operto
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridFrance
| | - Carles Falcon
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)MadridSpain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
| | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridFrance
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridFrance
| | - Luigi Lorenzini
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamNetherlands
| | - Silvia Ingala
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamNetherlands
| | - Alle Meije Wink
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamNetherlands
| | - Henk JMM Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamNetherlands
| | - Carolina Minguillón
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridFrance
| | - Karine Fauria
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridFrance
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
| | - Sven Haller
- CIRD Centre d'Imagerie Rive DroiteGenevaSwitzerland
| | - Gael Chetelat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and BrainCyceronFrance
| | - Adam Waldman
- Centre for Dementia Prevention, Edinburgh Imaging, and UK Dementia Research Institute at The University of EdinburghEdinburghUnited Kingdom
| | | | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamNetherlands
- Institutes of Neurology and Healthcare Engineering, University College LondonLondonUnited Kingdom
| | | | | | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, University of GothenburgMölndalSweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyLondonUnited Kingdom
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, University of GothenburgMölndalSweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University HospitalMölndalSweden
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridFrance
- Servei de Neurologia, Hospital del MarBarcelonaSpain
| | - Verónica Vilaplana
- Department of Signal Theory and Communications, Universitat Politècnica de CatalunyaBarcelonaSpain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center, Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)MadridSpain
| | | | | | | | | |
Collapse
|
25
|
Abu-Rumeileh S, Abdelhak A, Foschi M, D'Anna L, Russo M, Steinacker P, Kuhle J, Tumani H, Blennow K, Otto M. The multifaceted role of neurofilament light chain protein in non-primary neurological diseases. Brain 2023; 146:421-437. [PMID: 36083979 PMCID: PMC9494370 DOI: 10.1093/brain/awac328] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
The advancing validation and exploitation of CSF and blood neurofilament light chain protein as a biomarker of neuroaxonal damage has deeply changed the current diagnostic and prognostic approach to neurological diseases. Further, recent studies have provided evidence of potential new applications of this biomarker also in non-primary neurological diseases. In the present review we summarize the state of the art, future perspectives, but also limitations, of neurofilament light chain protein as a CSF and blood biomarker in several medical fields, including intensive care medicine, surgery, internal medicine and psychiatry. In particular, neurofilament light chain protein is associated with the degree of neurological impairment and outcome in patients admitted to intensive care units or in the perioperative phase and it seems to be highly interconnected with cardiovascular risk factors. Beyond that, interesting diagnostic and prognostic insights have been provided by the investigation of neurofilament light chain protein in psychiatric disorders as well as in the current coronavirus disease-19 pandemic and in normal ageing. Altogether, current data outline a multifaceted applicability of CSF and blood neurofilament light chain protein ranging from the critical clinical setting to the development of precision medicine models suggesting a strict interplay between the nervous system pathophysiology and the health-illness continuum.
Collapse
Affiliation(s)
- Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Ahmed Abdelhak
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, USA
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Matteo Foschi
- Department of Neuroscience, Neurology Unit – S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna, Italy
| | - Lucio D'Anna
- Department of Stroke and Neuroscience, Charing Cross Hospital, Imperial College London, NHS Healthcare Trust, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Michele Russo
- Department of Cardiology, S. Maria dei Battuti Hospital, AULSS 2 Veneto, Conegliano, Italy
| | - Petra Steinacker
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - 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
| | - Markus Otto
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
26
|
Amrein M, Meier S, Schäfer I, Schaedelin S, Willemse E, Benkert P, Walter J, Puelacher C, Zimmermann T, Median D, Egli C, Leppert D, Twerenbold R, Zellweger M, Kuhle J, Mueller C. Serum neurofilament light chain in functionally relevant coronary artery disease and adverse cardiovascular outcomes. Biomarkers 2023; 28:341-351. [PMID: 36714921 DOI: 10.1080/1354750x.2023.2172211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background: Functionally relevant coronary artery disease (fCAD), causing symptoms of myocardial ischemia, can currently only be reliably detected with advanced cardiac imaging. Serum neurofilament light chain (sNfL) is a biomarker for neuro-axonal injury known to be elevated by cardiovascular (CV) risk factors and cerebrovascular small-vessel diseases. Due to their pathophysiological similarities with fCAD and the link to CV risk factors, we hypothesised that sNfL may have diagnostic and prognostic value for fCAD and adverse cardiovascular outcomes.Methods: Of the large prospective Basel VIII study (NCT01838148), 4'016 consecutive patients undergoing cardiac work-up for suspected fCAD were included (median age 68 years, 32.5% women, 46.9% with history of CAD). The presence of fCAD was adjudicated using myocardial perfusion imaging single-photon emission tomography (MPI-SPECT) and coronary angiography. sNfL was measured using a high-sensitive single-molecule array assay. All-cause and cardiovascular death, myocardial infarction (MI), and stroke/transient ischaemic attack (TIA) during 5-year follow-up were the prognostic endpoints.Results: The diagnostic accuracy of sNfL for fCAD as quantified by the area under the curve (AUC) was low (0.58, 95%CI 0.56-0.60). sNfL was strongly associated with age, renal dysfunction, and body mass index and was a strong and independent predictor of all-cause death, cardiovascular death, and stroke/TIA but not MI. Time-dependent AUC for cardiovascular-death at 1-year was 0.85, 95%CI 0.80-0.89, and 0.81, 95%CI 0.77-0.86 at 2-years.Conclusion: While sNfL concentrations did not show a diagnostic role for fCAD, in contrast, sNfL was a strong and independent predictor of cardiovascular outcomes, including all-cause death, cardiovascular death and stroke/TIA.
Collapse
Affiliation(s)
- Melissa Amrein
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stephanie Meier
- Multiple Sclerosis Centre, Neurology, Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland.,Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Ibrahim Schäfer
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sabine Schaedelin
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Eline Willemse
- Multiple Sclerosis Centre, Neurology, Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland.,Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Pascal Benkert
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Joan Walter
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Christian Puelacher
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tobias Zimmermann
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Median
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Caroline Egli
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - David Leppert
- Multiple Sclerosis Centre, Neurology, Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland.,Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Raphael Twerenbold
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland.,University Center of Cardiovascular Science & Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Michael Zellweger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Multiple Sclerosis Centre, Neurology, Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland.,Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Christian Mueller
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
27
|
Zhu JD, Tsai SJ, Lin CP, Lee YJ, Yang AC. Predicting aging trajectories of decline in brain volume, cortical thickness and fractional anisotropy in schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2023; 9:1. [PMID: 36596800 PMCID: PMC9810255 DOI: 10.1038/s41537-022-00325-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023]
Abstract
Brain-age prediction is a novel approach to assessing deviated brain aging trajectories in different diseases. However, most studies have used an average brain age gap (BAG) of individuals with schizophrenia of different illness durations for comparison with healthy participants. Therefore, this study investigated whether declined brain structures as reflected by BAGs may be present in schizophrenia in terms of brain volume, cortical thickness, and fractional anisotropy across different illness durations. We used brain volume, cortical thickness, and fractional anisotropy as features to train three models from the training dataset. Three models were applied to predict brain ages in the hold-out test and schizophrenia datasets and calculate BAGs. We divided the schizophrenia dataset into multiple groups based on the illness duration using a sliding time window approach for ANCOVA analysis. The brain volume and cortical thickness models revealed that, in comparison with healthy controls, individuals with schizophrenia had larger BAGs across different illness durations, whereas the BAG in terms of fractional anisotropy did not differ from that of healthy controls after disease onset. Moreover, the BAG at the initial stage of schizophrenia was the largest in the cortical thickness model. In contrast, the BAG from approximately two decades after disease onset was the largest in the brain volume model. Our findings suggest that schizophrenia differentially affects the decline of different brain structures during the disease course. Moreover, different trends of decline in thickness and volume-based measures suggest a differential decline in dimensions of brain structure throughout the course of schizophrenia.
Collapse
Grants
- This work was supported by grants from the National Science and Technology Council, Taiwan (grant number 110-2321-B-A49A-502 and 110-2628-B-A49A-509, and 110-2634-F-075-001 to Albert C. Yang). Dr. Albert C. Yang was also supported by the Mt. Jade Young Scholarship Award from the Ministry of Education, Taiwan, as well as Brain Research Center, National Yang Ming Chiao Tung University, and the Ministry of Education (Aim for the Top University Plan), Taipei, Taiwan.
- Mr. J. D. Zhu was supported by the scholarship (108-2926-I-010-001-MY4) from the National Science and Technology Council, Taiwan.
- This work was supported by grants from the National Science and Technology Council, Taiwan (grant number 110-2321-B-A49A-502 and 110-2628-B-A49A-509, and 110-2634-F-075-001 to S. J. Tsai).
Collapse
Affiliation(s)
- Jun-Ding Zhu
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ju Lee
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Albert C Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Digital Medicine and Smart Healthcare Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
28
|
Rother C, Uhlmann RE, Müller SA, Schelle J, Skodras A, Obermüller U, Häsler LM, Lambert M, Baumann F, Xu Y, Bergmann C, Salvadori G, Loos M, Brzak I, Shimshek D, Neumann U, Walker LC, Schultz SA, Chhatwal JP, Kaeser SA, Lichtenthaler SF, Staufenbiel M, Jucker M. Experimental evidence for temporal uncoupling of brain Aβ deposition and neurodegenerative sequelae. Nat Commun 2022; 13:7333. [PMID: 36443293 PMCID: PMC9705543 DOI: 10.1038/s41467-022-34538-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022] Open
Abstract
Brain Aβ deposition is a key early event in the pathogenesis of Alzheimer´s disease (AD), but the long presymptomatic phase and poor correlation between Aβ deposition and clinical symptoms remain puzzling. To elucidate the dependency of downstream pathologies on Aβ, we analyzed the trajectories of cerebral Aβ accumulation, Aβ seeding activity, and neurofilament light chain (NfL) in the CSF (a biomarker of neurodegeneration) in Aβ-precursor protein transgenic mice. We find that Aβ deposition increases linearly until it reaches an apparent plateau at a late age, while Aβ seeding activity increases more rapidly and reaches a plateau earlier, coinciding with the onset of a robust increase of CSF NfL. Short-term inhibition of Aβ generation in amyloid-laden mice reduced Aβ deposition and associated glial changes, but failed to reduce Aβ seeding activity, and CSF NfL continued to increase although at a slower pace. When short-term or long-term inhibition of Aβ generation was started at pre-amyloid stages, CSF NfL did not increase despite some Aβ deposition, microglial activation, and robust brain Aβ seeding activity. A dissociation of Aβ load and CSF NfL trajectories was also found in familial AD, consistent with the view that Aβ aggregation is not kinetically coupled to neurotoxicity. Rather, neurodegeneration starts when Aβ seeding activity is saturated and before Aβ deposition reaches critical (half-maximal) levels, a phenomenon reminiscent of the two pathogenic phases in prion disease.
Collapse
Affiliation(s)
- Christine Rother
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
- Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074, Tübingen, Germany
| | - Ruth E Uhlmann
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
- Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074, Tübingen, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Juliane Schelle
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Angelos Skodras
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Ulrike Obermüller
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Lisa M Häsler
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Marius Lambert
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Frank Baumann
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Ying Xu
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Carina Bergmann
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
- Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074, Tübingen, Germany
| | - Giulia Salvadori
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Maarten Loos
- Sylics (Synaptologics BV), 3721 MA, Bilthoven, The Netherlands
| | - Irena Brzak
- Novartis Institutes for Biomedical Research, CH-4056, Basel, Switzerland
| | - Derya Shimshek
- Novartis Institutes for Biomedical Research, CH-4056, Basel, Switzerland
| | - Ulf Neumann
- Novartis Institutes for Biomedical Research, CH-4056, Basel, Switzerland
| | - Lary C Walker
- Department of Neurology and Emory National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
| | | | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Stephan A Kaeser
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Matthias Staufenbiel
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany.
| |
Collapse
|
29
|
Mengel-From J, Nielsen KE, Clemmensen SB, Feddersen S, Christensen K, Hjelmborg JVB, Tan Q, Nygaard M. Circulating microRNAs related to lipid metabolism and solid tissue maintenance and morphology associate with mortality in elderly twins. Exp Gerontol 2022; 169:111980. [PMID: 36244587 DOI: 10.1016/j.exger.2022.111980] [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: 07/01/2022] [Revised: 09/23/2022] [Accepted: 10/08/2022] [Indexed: 12/15/2022]
Abstract
The lifespan of humans varies greatly between individuals. Here, we aimed to explore what biological roles miRNAs may have on old age mortality-variation. Circulating miRNAs were measured in plasma from 43 monozygotic twin pairs (73-95 years of age) and mortality analyses were applied using Cox regression survival analyses and linear regression analyses of lifespan. In general, nominally significant miRNAs were mainly upregulated with shorter lifespan, both in Cox analysis (72 % upregulated) and in linear regression analysis (81 % upregulated). A total of 29 miRNAs were associated to mortality at a nominal significance level (p < 0.05) in the survival analysis, but no miRNAs passed the FDR adjusted level of significance. Seven of the 29 miRNAs; hsa-miR-140-3p, hsa-miR-16-5p, hsa-miR-487b-3p, hsa-miR-19a-3p, hsa-let-7d-5p, hsa-miR-320a, hsa-miR-375, were nominally significant across two linear twin-paired analyses and the cox analysis. Pathway analyses of the 29 nominally significant miRNAs from the individual level analyses resulted in two nominally significant associated Reactome pathways (unadjusted p < 0.05); 'Negative regulation of FGFR signaling' and 'Neurotransmitter receptor binding and downstream transmission in the postsynaptic cell', and two significantly associated KEGG pathways; 'Linoleic acid metabolism' and 'Toxoplasmosis'. Additional pathway analyses and results of previous studies support that miRNAs linked to mortality at age 70 years or older play a role in lipid metabolism, tissues maintenance and morphology.
Collapse
Affiliation(s)
- Jonas Mengel-From
- The Danish Aging Research Center and Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Department of Clinical Genetics, Odense University Hospital, 5000 Odense, Denmark.
| | - Katrine Egebjerg Nielsen
- The Danish Aging Research Center and Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, 5000 Odense, Denmark
| | - Signe Bedsted Clemmensen
- The Danish Aging Research Center and Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, 5000 Odense, Denmark
| | - Søren Feddersen
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Kaare Christensen
- The Danish Aging Research Center and Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Department of Clinical Genetics, Odense University Hospital, 5000 Odense, Denmark; Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Jacob V B Hjelmborg
- The Danish Aging Research Center and Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, 5000 Odense, Denmark
| | - Qihua Tan
- The Danish Aging Research Center and Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Department of Clinical Genetics, Odense University Hospital, 5000 Odense, Denmark
| | - Marianne Nygaard
- The Danish Aging Research Center and Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Department of Clinical Genetics, Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
30
|
Nguyen AD, Malmstrom TK, Aggarwal G, Miller DK, Vellas B, Morley JE. Serum neurofilament light levels are predictive of all-cause mortality in late middle-aged individuals. EBioMedicine 2022; 82:104146. [PMID: 35830835 PMCID: PMC9284367 DOI: 10.1016/j.ebiom.2022.104146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/13/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Blood biomarkers can offer valuable and easily accessible indicators of normal biological processes, pathogenic conditions, and responses to therapeutic interventions. Recent studies found that levels of neurofilament light chain (NfL) in the blood are associated with mortality in three European cohorts of older adults (median ages 73, 93, and 100 years). Whether similar associations exist in younger adults and in other ethnic groups is currently not known. METHODS We utilized a cohort study that included 294 African Americans (baseline ages 49-65). Serum NfL levels were measured using a Meso Scale Discovery-based assay. Vital status was determined by matching through the National Death Index. FINDINGS Seventy-two participants (24.5%) died during the 14-15 years of follow up (2000-2014). Baseline serum NfL levels were significantly higher in the decedent group (86.1±65.7 pg/ml vs. 50.1±28.0 pg/ml, p < 0·001). In binomial logistic regression models adjusted for age, gender, education, baseline smoking status, BMI, and total comorbidities (0-11), serum NfL levels remained a strong predictor of all-cause mortality, and sensitivity analyses employing multiple additional covariates did not substantively change the relationship. Further, Kaplan-Meier curves based on serum NfL quartiles showed reduced survival in groups with higher serum NfL levels. INTERPRETATION This study found a positive association between serum NfL levels and mortality in late middle-aged and older individuals. While our findings support that serum NfL levels may be a useful biomarker for all-cause mortality, further studies are needed to understand the biological mechanisms underlying this association. FUNDING National Institute on Aging, Saint Louis University.
Collapse
Affiliation(s)
- Andrew D Nguyen
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA.
| | - Theodore K Malmstrom
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA; Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Geetika Aggarwal
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA
| | | | - Bruno Vellas
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France; UMR 1295 INSERM, University of Toulouse III, Toulouse, France
| | - John E Morley
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
31
|
Ali SK, Ali RH. Effects of antidiabetic agents on Alzheimer's disease biomarkers in experimentally induced hyperglycemic rat model by streptozocin. PLoS One 2022; 17:e0271138. [PMID: 35802659 PMCID: PMC9269384 DOI: 10.1371/journal.pone.0271138] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Alzheimer's disease is the most common cause of dementia in the elderly population. It is characterized by the accumulation of amyloid β and intraneuronal neurofibrillary tangles in the brain. Increasing evidence shows that the disturbance of insulin signalling in the brain may contribute to the pathophysiology of Alzheimer's disease. In type 1 diabetes, these disruptions are caused by hypoinsulinemia, but in type 2 diabetes, they are caused by insulin resistance and decreased insulin secretion. Multiple studies have shown that diabetes is connected with an increased risk of acquiring Alzheimer's disease. The aim of this study was to investigate the impact of anti-diabetic agents on Alzheimer's disease progression and the levels of Alzheimer's biomarkers in a hyperglycaemic rat model, which was induced by intraperitoneal injection of streptozocin to produce insulin-deficient diabetes. METHOD Thirty-six male Wistar albino rats were allocated into six groups of six rats each. Group I was the negative control group. Intraperitoneal injections of streptozocin (42mg/kg) were used once for the five experimental groups. Group II served as the positive control group. The rats in Groups III, IV, V, and VI received metformin (300mg/kg), donepezil (10mg/kg), insulin glargine (3 unit/animal), and glibenclamide (10mg/kg), respectively, for 21 days. RESULTS Inducing hyperglycaemia in rats significantly increased the levels of serum glucose, haemoglobin A1c, total cholesterol, triglycerides, high-density lipoprotein, interleukin 6, tumour necrosis factor alpha, amyloid β 42, total plasma tau, and neurofilament light. A significant increase was also found in brain amyloid β 42, nitric oxide, acetylcholinesterase, malondialdehyde, β secretase, and phosphorylated microtubule-associated protein tau. The greatest statistically significant reductions in serum glucose, haemoglobin A1c, triglycerides, amyloid β 42, total plasma tau, brain amyloid β 42, acetylcholinesterase, and malondialdehyde were observed in rats treated with metformin. In contrast, rats treated with donepezil demonstrated the greatest statistically significant reduction in serum tumour necrosis factor alpha, brain nitric oxide, and β secretase. The levels of neurofilament light and phosphorylated microtubule-associated protein tau in the brains of rats treated with insulin glargine were significantly lower than the other treatment groups. The total cholesterol and low-density lipoprotein levels in rats treated with glibenclamide exhibited the most statistically significant reductions of all the treatment groups. CONCLUSIONS Metformin and donepezil, when administered at appropriate doses, were shown to successfully lower most plasma and brain biomarkers, including glucose, triglycerides, tumour necrosis factor alpha, amyloid β 42, nitric oxide, acetylcholinesterase, malondialdehyde, and β secretase in rats suffering from Diabetes Mellitus. As a result of this research, we suggest that metformin, either alone or in conjunction with donepezil, might be an excellent drug of choice for neuro-regeneration and risk reduction in Alzheimer's like disease.
Collapse
Affiliation(s)
- Shatw Khalid Ali
- Department of Pharmacology and Toxicology, Hawler Medical University, Erbil, Iraq
| | - Rojgar H. Ali
- Department of Pharmacology and Toxicology, Hawler Medical University, Erbil, Iraq
| |
Collapse
|
32
|
Beydoun MA, Noren Hooten N, Weiss J, Beydoun HA, Hossain S, Evans MK, Zonderman AB. Plasma neurofilament light and its association with all-cause mortality risk among urban middle-aged men and women. BMC Med 2022; 20:218. [PMID: 35692046 PMCID: PMC9190073 DOI: 10.1186/s12916-022-02425-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/31/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Neurofilament light chain (NfL) is released into the blood during neuronal damage. NfL is linked to mortality in neurological disorders, remaining unexplored in population studies. We investigated whether initial (v1) and annualized change (δ) in plasma NfL can predict all-cause mortality in middle-aged dementia-free urban adults. METHODS Longitudinal data were from 694 participants in the Healthy Aging in Neighborhoods of Diversity Across the Life Span study (HANDLS, mean agev1: 47.8 years, 42% male, 55.8% African American). Plasma NfL was measured prospectively at three visits. Analyses included Cox proportional hazards models for all-cause mortality risk and 4-way decomposition testing for interaction and mediation. RESULTS Unlike men, women exhibited a direct association between δNfL (above vs. below median) and all-cause mortality risk in both the minimally (HR = 3.91, 95% CI 1.10-13.9, p = 0.036) and fully adjusted models (HR = 4.92, 95% CI 1.26-19.2, p = 0.022), and for δNfL (per unit increase) in the full model (HR = 1.65, 95% CI 1.04-2.61, p = 0.034). In both models, and among women, 1 standard deviation of NfLv1 was associated with an increased all-cause mortality risk (reduced model: HR = 2.01, 95% CI 1.24-3.25, p = 0.005; full model: HR = 1.75, 95% CI 1.02-2.98, p = 0.041). Only few interactions were detected for cardio-metabolic risk factors. Notably, NfLv1 was shown to be a better prognostic indicator at normal hsCRP values among women, while HbA1c and δNfL interacted synergistically to determine mortality risk, overall. CONCLUSIONS These findings indicate that plasma NfL levels at baseline and over time can predict all-cause mortality in women and interacts with hsCRP and HbA1c to predict that risk.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA.
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Jordan Weiss
- Department of Demography, University of California, Berkeley, Berkeley, CA, USA
| | - Hind A Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA, USA
| | - Sharmin Hossain
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| |
Collapse
|
33
|
Lu WH, Giudici KV, Morley JE, Guyonnet S, Parini A, Aggarwal G, Nguyen AD, Li Y, Bateman RJ, Vellas B, de Souto Barreto P. Investigating the combination of plasma amyloid-beta and geroscience biomarkers on the incidence of clinically meaningful cognitive decline in older adults. GeroScience 2022; 44:1489-1503. [PMID: 35445358 PMCID: PMC9213609 DOI: 10.1007/s11357-022-00554-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/22/2022] [Indexed: 11/04/2022] Open
Abstract
We investigated combining a core AD neuropathology measure (plasma amyloid-beta [Aβ] 42/40) with five plasma markers of inflammation, cellular stress, and neurodegeneration to predict cognitive decline. Among 401 participants free of dementia (median [IQR] age, 76 [73-80] years) from the Multidomain Alzheimer Preventive Trial (MAPT), 28 (7.0%) participants developed dementia, and 137 (34.2%) had worsening of clinical dementia rating (CDR) scale over 4 years. In the models utilizing plasma Aβ alone, a tenfold increased risk of incident dementia (nonsignificant) and a fivefold increased risk of worsening CDR were observed as each nature log unit increased in plasma Aβ levels. Models incorporating Aβ plus multiple plasma biomarkers performed similarly to models included Aβ alone in predicting dementia and CDR progression. However, improving Aβ model performance for composite cognitive score (CCS) decline, a proxy of dementia, was observed after including plasma monocyte chemoattractant protein 1 (MCP1) and growth differentiation factor 15 (GDF15) as covariates. Participants with abnormal Aβ, GDF15, and MCP1 presented higher CCS decline (worsening cognitive function) compared to their normal-biomarker counterparts (adjusted β [95% CI], - 0.21 [- 0.35 to - 0.06], p = 0.005). In conclusion, our study found limited added values of multi-biomarkers beyond the basic Aβ models for predicting clinically meaningful cognitive decline among non-demented older adults. However, a combined assessment of inflammatory and cellular stress status with Aβ pathology through measuring plasma biomarkers may improve the evaluation of cognitive performance.
Collapse
Affiliation(s)
- Wan-Hsuan Lu
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
- Inserm CERPOP - UMR1295, University of Toulouse III, 31000, Toulouse, France
| | - Kelly Virecoulon Giudici
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
| | - John E Morley
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Sophie Guyonnet
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
- Inserm CERPOP - UMR1295, University of Toulouse III, 31000, Toulouse, France
| | - Angelo Parini
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Inserm UMR 1048, University of Toulouse, 31400, Toulouse, France
| | - Geetika Aggarwal
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
- Inserm CERPOP - UMR1295, University of Toulouse III, 31000, Toulouse, France
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), 31000, Toulouse, France
- Inserm CERPOP - UMR1295, University of Toulouse III, 31000, Toulouse, France
| |
Collapse
|
34
|
O'Bryant S, Petersen M, Hall J, Johnson L, Yaffe K, Braskie M, Toga AW, Rissman RA. Characterizing plasma NfL in a community-dwelling multi-ethnic cohort: Results from the HABLE study. Alzheimers Dement 2022; 18:240-250. [PMID: 34310015 PMCID: PMC9228481 DOI: 10.1002/alz.12404] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 01/27/2023]
Abstract
INTRODUCTION No large-scale characterizations of neurofilament light chain (NfL) have been conducted in diverse populations. METHODS Baseline data were analyzed among n = 890 Mexican Americans and n = 813 non-Hispanic Whites from the multi-ethnic Health & Aging Brain among Latino Elders (HABLE) study. Plasma NfL was measured on the Simoa platform. RESULTS In unadjusted models, NfL was significantly associated with age (P < .001), hypertension (P < .001), dyslipidemia (P = .02), and diabetes (P < .001). Covarying for age and sex, NfL was associated with neurodegeneration (P < .001) and global amyloid burden levels (P = .02) in a subset with available data. NfL levels were significantly associated with diagnostic groups (Normal Cognition [NC], mild cognitive impairment [MCI], Dementia; P < .001); however, there was no cut-score that yielded acceptable diagnostic accuracy. NfL levels produced a sensitivity of 0.60 and specificity of 0.78 with negative predictive value of 89% for detecting amyloid positivity. DISCUSSION Plasma NfL levels are significantly impacted by age and medical co-morbidities that are common among older adults, which complicate its utility as a diagnostic biomarker.
Collapse
Affiliation(s)
- Sid O'Bryant
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Melissa Petersen
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
- Department of Family MedicineUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - James Hall
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
- Department of Pharmacology and NeuroscienceUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Leigh Johnson
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
- Department of Pharmacology and NeuroscienceUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Kristine Yaffe
- Department of PsychiatryNeurology, and Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCaliforniaUSA
- San Francisco VA Medical CenterSan FranciscoCaliforniaUSA
| | - Meredith Braskie
- Imaging Genetics CenterStevens Neuroimaging and Informatics InstituteKeck School of MedicineUSCLos AngelesCaliforniaUSA
| | - Arthur W. Toga
- Laboratory of Neuro ImagingUSC Stevens Neuroimaging and Informatics InstituteKeck School of Medicine of USCUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Robert A. Rissman
- Department of NeurosciencesUniversity of CaliforniaSan DiegoLa JollaCaliforniaUSA
- Veterans Affairs San Diego Healthcare SystemSan DiegoCaliforniaUSA
| | | |
Collapse
|
35
|
Halbgebauer S, Steinacker P, Verde F, Weishaupt J, Oeckl P, von Arnim C, Dorst J, Feneberg E, Mayer B, Rosenbohm A, Silani V, Ludolph AC, Otto M. Comparison of CSF and serum neurofilament light and heavy chain as differential diagnostic biomarkers for ALS. J Neurol Neurosurg Psychiatry 2022; 93:68-74. [PMID: 34417339 DOI: 10.1136/jnnp-2021-327129] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Elevated levels of neurofilament light (NfL) and heavy (NfH) chain in amyotrophic lateral sclerosis (ALS) cerebrospinal fluid (CSF) and serum reflect neuro-axonal degeneration and are used as diagnostic biomarkers. However, studies comparing the differential diagnostic potential for ALS of all four parameters are missing. Here, we measured serum NfL/NfH and CSF NfL/NfH in a large cohort of ALS and other neurological disorders and analysed the differential diagnostic potential. METHODS In total CSF and serum of 294 patients were analysed. The diagnostic groups comprised: ALS (n=75), frontotemporal lobar degeneration (FTLD) (n=33), Alzheimer's disease (n=20), Parkinson's disease (dementia) (n=18), Creutzfeldt-Jakob disease (n=11), non-neurodegenerative controls (n=77) (Con) and 60 patients who were seen under the direct differential diagnosis of a patient with ALS (Con.DD). RESULTS CSF and serum NfL and NfH showed significantly increased levels in ALS (p<0.0001) compared with Con and Con.DD. The difference between ALS and FTLD was markedly stronger for NfH than for NfL. CSF and serum NfL demonstrated a stronger correlation (r=0.84 (95% CI 0.80 to 0.87), p<0.001) than CSF and serum NfH (r=0.68 (95% CI 0.61 to 0.75), p<0.0001). Comparing ALS and Con.DD, receiver operating characteristic analysis revealed the best area under the curve (AUC) value for CSF NfL (AUC=0.94, 95% CI 0.91 to 0.98), followed by CSF NfH (0.93, 95% CI 0.88 to 0.98), serum NfL (0.93, 95% CI 0.89 to 0.97) and serum NfH (0.88, 95% CI 0.82 to 0.94). CONCLUSION Our results demonstrate that CSF NfL and NfH as well as serum NfL are equally suited for the differential diagnosis of ALS, whereas serum NfH appears to be slightly less potent.
Collapse
Affiliation(s)
| | | | - Federico Verde
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy.,Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milano, Italy
| | - Jochen Weishaupt
- Department of Neurology, Institute for Neurodegeneration, Universitätsmedizin Mannheim, Mannheim, Germany
| | - Patrick Oeckl
- Neurology, University of Ulm, Ulm, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE e.V.), Ulm, Germany
| | | | | | - Emily Feneberg
- Department of Neurology, University Hospital Rechts der Isar, Munich, Bayern, Germany
| | - Benjamin Mayer
- Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | | | - Vincenzo Silani
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy.,Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milano, Italy
| | | | - Markus Otto
- Neurology, University of Ulm, Ulm, Germany .,Department of Neurology, University clinic, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
36
|
Anisman H, Kusnecov AW. Stress, immunity, and cancer. Cancer 2022. [DOI: 10.1016/b978-0-323-91904-3.00017-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
37
|
Koini M, Pirpamer L, Hofer E, Buchmann A, Pinter D, Ropele S, Enzinger C, Benkert P, Leppert D, Kuhle J, Schmidt R, Khalil M. Factors influencing serum neurofilament light chain levels in normal aging. Aging (Albany NY) 2021; 13:25729-25738. [PMID: 34923481 PMCID: PMC8751593 DOI: 10.18632/aging.203790] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/08/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Serum neurofilament light (sNfL) is a promising marker for neuro-axonal damage and it is now well known that its levels also increase with higher age. However, the effect of other determinants besides age is still poorly investigated. We therefore aimed to identify factors influencing the sNfL concentration by analysing a large set of demographical, life-style and clinical variables in a normal aging cohort. METHODS sNfL was quantified by single molecule array (Simoa) assay in 327 neurologically inconspicuous individuals (median age 67.8±10.7 years, 192 female) who participated in the Austrian Stroke Prevention Family Study (ASPS-Fam). Random forest regression analysis was used to rank the association of included variables with sNfL in the entire cohort and in age-stratified subgroups. Linear regression then served to identify factors independently influencing sNfL concentration. RESULTS Age (β=0.513, p<0.001) was by far the most important factor influencing sNfL, which was mainly driven by individuals ≥60 years. In age stratified sub-groups, body mass index (BMI) (β=-0.298, p<0.001) independently predicted sNfL in individuals aged 38-60 years. In individuals ≥60 years, age (β=0.394, p<0.001), renal function (β=0.376, p<0.001), blood volume (β=-0.198, p=0.008) and high density lipoprotein (HDL) (β=0.149, p=0.013) were associated with sNfL levels. CONCLUSIONS Age is the most important factor influencing sNfL concentrations, getting increasingly relevant in elderly people. BMI further influences sNfL levels, especially at younger age, whereas renal function gets increasingly relevant in the elderly.
Collapse
Affiliation(s)
- Marisa Koini
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Lukas Pirpamer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | | | - Daniela Pinter
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Christian Enzinger
- Department of Neurology, Medical University of Graz, Graz, Austria
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Pascal Benkert
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - David Leppert
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria
| |
Collapse
|
38
|
Lu WH, Giudici KV, Guyonnet S, Aggarwal G, Nguyen AD, Morley JE, Vellas B, de Souto Barreto P. Associations of plasma neurofilament light chain and progranulin with frailty in older adults. J Am Geriatr Soc 2021; 70:1236-1243. [PMID: 34908159 DOI: 10.1111/jgs.17604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/16/2021] [Accepted: 11/21/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND In previous studies, plasma neurofilament light chain (NfL) and progranulin (PGRN) levels are associated with cognitive and physical impairment in older individuals. However, evidence of their relationships with frailty is lacking. This study aims to explore the associations of plasma NfL and PGRN levels with frailty in community-dwelling older adults. METHODS We included 507 older adults (mean [standard deviation] age, 76.7 [4.5] years) with plasma NfL and PGRN measurements from the Multidomain Alzheimer Preventive Trial (MAPT). The timepoint of biomarker measurements, either 12 or 24 months after study enrollment, was defined as the baseline for each participant. Frailty phenotype (robust, pre-frail, and frail) was assessed at 12, 24, 36, 48, and 60 months by Fried's frailty criteria. The cross-sectional associations between plasma neurodegenerative biomarkers and frailty severity were examined using logistic regressions. We further used Cox proportional hazard models to evaluate the associations between plasma biomarkers and incident frailty among robust or pre-frail participants at baseline (n = 403). RESULTS At baseline, participants with high plasma NfL levels (>93.11 pg/ml [the upper quartile]) had a higher likelihood of pre-frailty or frailty compared to their normal NfL counterparts (odds ratio = 1.68; 95% confidence interval = 1.10-2.57); however, this association did not remain significant after controlling for covariates. Neither NfL nor PGRN levels showed prospective associations with incident frailty over 4 years. CONCLUSIONS This study failed to find associations of circulating NfL and PGRN levels with frailty among community-dwelling older adults in adjusted analyses. Whether plasma neurodegenerative markers serve as potential biomarkers of frailty requires further investigation.
Collapse
Affiliation(s)
- Wan-Hsuan Lu
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France.,Maintain Aging Research Team, CERPOP, Inserm, Université Paul Sabatier, Toulouse, France
| | - Kelly Virecoulon Giudici
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France
| | - Sophie Guyonnet
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France.,Maintain Aging Research Team, CERPOP, Inserm, Université Paul Sabatier, Toulouse, France
| | - Geetika Aggarwal
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri, USA
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri, USA
| | - John E Morley
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France.,Maintain Aging Research Team, CERPOP, Inserm, Université Paul Sabatier, Toulouse, France
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France.,Maintain Aging Research Team, CERPOP, Inserm, Université Paul Sabatier, Toulouse, France
| | | |
Collapse
|