RT Journal Article SR Electronic T1 Shorter leukocyte telomere length is associated with distinct CSF biomarker dynamics across early AD stages in at-risk individuals JF medRxiv FD Cold Spring Harbor Laboratory Press SP 2024.12.03.24318248 DO 10.1101/2024.12.03.24318248 A1 Rodríguez-Fernández, Blanca A1 González-Escalante, Armand A1 Genius, Patricia A1 Evans, Tavia A1 Ortiz-Romero, Paula A1 Minguillón, Carolina A1 Kollmorgen, Gwendlyn A1 Ashton, Nicholas A. A1 Zetterberg, Henrik A1 Blennow, Kaj A1 Gispert, Juan Domingo A1 Navarro, Arcadi A1 Suárez-Calvet, Marc A1 Sala-Vila, Aleix A1 Crous-Bou, Marta A1 Vilor-Tejedor, Natàlia A1 ALFA study YR 2024 UL http://medrxiv.org/content/early/2024/12/05/2024.12.03.24318248.abstract AB INTRODUCTION Telomere length (TL) is a hallmark of biological aging. Shorter TL has been linked to an increased risk of Alzheimer’s disease (AD), but its role in AD pathophysiology remains unclear. This study investigates the relationship between TL, longitudinal cerebrospinal fluid (CSF) AD biomarkers, and brain structure in cognitively unimpaired (CU) individuals at risk for AD.METHODS We analyzed data from 346 middle-aged CU ALFA+ participants, measuring leukocyte TL (LTL) by qPCR. AD-related CSF biomarkers were measured at baseline and after 3 years. Stratified analyses by APOE-e4 and amyloid-tau (AT) status were conducted.RESULTS Shorter LTL was associated with higher astrocytic reactivity and synaptic dysfunction biomarkers, as well as thicker cortex in AD-vulnerable regions. Astrocytic biomarkers mediated the LTL-cortical thickness association. In APOE-e4 carriers and AT-positive individuals, shorter LTL linked to higher p-tau181 and neurodegeneration markers.CONCLUSION These findings highlight telomere shortening as a potential contributor of early AD-related progression.HighlightsShorter leukocyte telomere length (LTL) was associated with higher levels of cerebrospinal fluid (CSF) GFAP, CSF S100B and CSF α-synuclein, independently of amyloid and tau pathology.Shorter LTL was associated with higher baseline CSF NfL and t-tau levels in the A+T- and A+T+ groups, respectively.LTL association with brain structure was partially mediated by CSF biomarkers of astrocytic reactivity.Research in context1. Systematic review: Literature review was performed using traditional sources (e.g., PubMed). While the association between leukocyte telomere length (LTL) shortening and increased sporadic AD risk is well-documented, its role in AD pathogenesis remains unclear. These findings have been appropriately referenced.2. Interpretation: In cognitively unimpaired adults at higher risk for AD, shorter LTL was associated with high AD-related cerebrospinal fluid (CSF) biomarkers, including p-tau181 and biomarkers of neurodegeneration, synaptic dysfunction, glial reactivity, and inflammation. These associations were either more pronounce or exclusively observed in APOE-e4 carriers and individuals with early AD pathology (measured by CSF Aβ42/40 and p-tau181). Furthermore, increased astrocytic reactivity mediated the relationship between LTL and brain structure integrity.3. Future directions: Further research is needed to understand the role of peripheral aging in AD pathology. Investigating how peripheral immune aging influences brain homeostasis and AD progression could help identify early targets of neuroinflammation and neurodegeneration.Competing Interest StatementBRF, AGE, TEE, MMA, MS, POR, CM, NVT, NA, ANC, MCB, and ASV have nothing to disclose. GSB has served as a consultant for Roche Farma, S.A. GK is a full-time employee of Roche Diagnostics GmbH. CQ-R is a full-time employee of Roche Diagnostics International Ltd. JDG receives research funding from Roche Diagnostics, Hoffmann-La Roche, and GE Healthcare; has given lectures in symposia sponsored by Biogen, Philips, and Life-MI; and consulting fees from Roche Diagnostics and serves in a scientific advisory board at Prothena Biosciences. HZ has served at scientific advisory boards and/or as a consultant for Abbvie, Acumen, Alector, Alzinova, ALZpath, Amylyx, Annexon, Apellis, Artery Therapeutics, AZTherapies, Cognito Therapeutics, CogRx, Denali, Eisai, LabCorp, Merry Life, Nervgen, Novo Nordisk, Optoceutics, Passage Bio, Pinteon Therapeutics, Prothena, Quanterix, Red Abbey Labs, reMYND, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics, and Wave, has given lectures sponsored by Alzecure, BioArctic, Biogen, Cellectricon, Fujirebio, Lilly, Novo Nordisk, Roche, and WebMD, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside submitted work). KB has served as a consultant, on advisory boards, or on data monitoring committees for Abcam, Axon, Biogen, JOMDD/Shimadzu, Julius Clinical, Lilly, MagQu, Novartis, Prothena, Roche Diagnostics, and Siemens Healthineers, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program. Author disclosures are available in the supporting information. MS-C has received in the past 36mo consultancy/speaker fees (paid to the institution) from by Almirall, Eli Lilly, Novo Nordisk, and Roche Diagnostics. He has received consultancy fees or served on advisory boards (paid to the institution) of Eli Lilly, Grifols and Roche Diagnostics. He was granted a project and is a site investigator of a clinical trial (funded to the institution) by Roche Diagnostics. In-kind support for research (to the institution) was received from ADx Neurosciences, Alamar Biosciences, ALZPath, Avid Radiopharmaceuticals, Eli Lilly, Fujirebio, Janssen Research & Development, Meso Scale Discovery, and Roche Diagnostics; MS-C did not receive any personal compensation from these organizations or any other for-profit organization.Funding StatementThe project leading to these results has received funding from the Alzheimers Association (Grant AARG–19–618265). This project has received funding from Instituto de Salud Carlos III (PI19/00119). Additional support has been received from la Caixa Foundation (ID 100010434), under agreement LCF/PR/GN17/50300004, the Alzheimers Association and an international anonymous charity foundation through the TriBEKa Imaging Platform project (TriBEKa–17–519007), the Health Department of the Catalan Government (Health Research and Innovation Strategic Plan (PERIS) 2016–2020 grant# SLT002/16/00201), and the Universities and Research Secretariat, Ministry of Business and Knowledge of the Catalan Government under the grant no. 2017–SGR–892. All CRG authors acknowledge the support of the Spanish Ministry of Science, Innovation, and Universities to the EMBL partnership, the Centro de Excelencia Severo Ochoa, and the CERCA Programme / Generalitat de Catalunya. JDG is supported by the Spanish Ministry of Science and Innovation (RYC–2013–13054). GSB has received funding from the Spanish Ministry of Science and Innovation, Spanish Research Agency (PID2020–119556RA–I00). NV–T was supported by the Spanish Ministry of Science and Innovation – State Research Agency IJC2020–043216–IMCINAEI101303950110033 and the European Union NextGenerationEU/PRTR and currently receives funding from the Spanish Research Agency MICIUAEI101303950110033 grant RYC2022–038136–I cofunded by the European Union FSE+ and grant PID2022–143106OA–I00 cofunded by the European Union FEDER. Additionally, NV–T is supported in part by the William H Gates Sr Fellowship from the Alzheimers Disease Data Initiative.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:The ALFA+ study was approved by the independent ethics committee of Parc de Salut Mar Barcelona and has been registered as a clinical trial (identifier: NCT02485730). All study participants provided written informed consent for study participation.I confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines, such as any relevant EQUATOR Network research reporting checklist(s) and other pertinent material, if applicable.YesAll data produced in the present study are available upon reasonable request to the authors.Aβamyloid-βADAlzheimer’s diseaseATamyloid β-tau stagingCAIDECardiovascular Risk Factors, Aging, and Incidence of DementiaCDRClinical Dementia RatingCSFcerebrospinal fluidCUcognitively unimpairedCVcoefficient of variationDNAdeoxyribonucleic acidFDRFalse Discovery RateGFAPglial fibrillary acidic proteinIL-6interleukin 6LTLleukocyte telomere lengthMCImild cognitive impairmentMMSEMini-Mental State ExaminationMRImagnetic resonance imagingNfLneurofilament lightPETpositron emission tomographyp-tau181phosphorylated tau181RT-qPCRreal-time polymerase chain reaction-based assaysTREM2soluble triggering receptor expressed on myeloid cells 2TLtelomere lengtht-tautotal tauYKL-40chitinase-3-like protein 1