1
|
Milos T, Vuic B, Balic N, Farkas V, Nedic Erjavec G, Svob Strac D, Nikolac Perkovic M, Pivac N. Cerebrospinal fluid in the differential diagnosis of Alzheimer's disease: an update of the literature. Expert Rev Neurother 2024; 24:1063-1079. [PMID: 39233323 DOI: 10.1080/14737175.2024.2400683] [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: 04/23/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The importance of cerebrospinal fluid (CSF) biomarkers in Alzheimer's disease (AD) diagnosis is rapidly increasing, and there is a growing interest in the use of CSF biomarkers in monitoring the response to therapy, especially in the light of newly available approaches to the therapy of neurodegenerative diseases. AREAS COVERED In this review we discuss the most relevant measures of neurodegeneration that are being used to distinguish patients with AD from healthy controls and individuals with mild cognitive impairment, in order to provide an overview of the latest information available in the scientific literature. We focus on markers related to amyloid processing, markers associated with neurofibrillary tangles, neuroinflammation, neuroaxonal injury and degeneration, synaptic loss and dysfunction, and markers of α-synuclein pathology. EXPERT OPINION In addition to neuropsychological evaluation, core CSF biomarkers (Aβ42, t-tau, and p-tau181) have been recommended for improvement of timely, accurate and differential diagnosis of AD, as well as to assess the risk and rate of disease progression. In addition to the core CSF biomarkers, various other markers related to synaptic dysfunction, neuroinflammation, and glial activation (neurogranin, SNAP-25, Nfl, YKL-40, TREM2) are now investigated and have yet to be validated for future potential clinical use in AD diagnosis.
Collapse
Affiliation(s)
- Tina Milos
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Barbara Vuic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Nikola Balic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Vladimir Farkas
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | | | | | | | - Nela Pivac
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
- University of Applied Sciences Hrvatsko Zagorje Krapina, Krapina, Croatia
| |
Collapse
|
2
|
Li J, Liu PP, Wang Y, Ren CY, Zhang M. Lectin YKL-40 Level and Telomere Length are Indicators of Insomnia Disorder. J Integr Neurosci 2024; 23:180. [PMID: 39344239 DOI: 10.31083/j.jin2309180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 10/01/2024] Open
Abstract
OBJECTIVE To explore the relationship between YKL-40 level, telomere length, and different subtypes of insomnia disorder. METHODS A total of 145 individuals suffering from insomnia were enrolled and divided into four groups according to the insomniac subtypes: difficulty initiating sleep, early morning awakening, difficulty maintaining sleep, and mixed symptoms. Eighty healthy controls were also collected at the same time. Peripheral leukocyte genomic DNA was extracted, relative telomere lengths were measured using the real-time quantitative polymerase chain reaction method, and YKL-40 levels were determined using enzyme-linked immunoassay. Logistic regression modeling was used to analyze the correlation between different insomnia subtypes, YKL-40 level, and telomere length. RESULTS People with telomere lengths in the lowest tertile were more likely to have trouble falling asleep (odds ratio (OR) 2.13, 95% confidence interval (CI) 1.22-3.63; p = 0.03) and had a higher frequency of mixed symptoms (OR 1.49, 95% CI 1.30-2.81; p = 0.04). People in the highest tertile of YKL-40 level had an increased chance of waking up early (OR 2.98, 95% CI 1.54-5.33; p = 0.01) and more mixed symptoms (OR 1.47, 95% CI 1.22-2.79; p = 0.02). Furthermore, using receiver operating characteristic curve analysis, the area under the curve of YKL-40 level and telomere length was 0.806 and 0.746, respectively. CONCLUSIONS Telomere length in patients with difficulty initiating sleep and mixed symptoms was significantly shortened and the level of YKL-40 in people who have early morning awakening and mixed symptoms was significantly increased. Our findings provide the first evidence that leukocyte telomere length and YKL-40 level are individually linked to mixed symptoms.
Collapse
Affiliation(s)
- Jing Li
- Department of Neurology, The First Hospital of Anhui University of Science and Technology (Huainan First People's Hospital), 232000 Huainan, Anhui, China
| | - Pei-Pei Liu
- Department of Neurology, Fu Yang Fifth People's Hospital, 236000 Fuyang, Anhui, China
| | - Yan Wang
- Department of Neurology, Hefei Hospital Affiliated to Anhui Medical University, The Second People's Hospital of Hefei, 230000 Hefei, Anhui, China
| | - Chong-Yang Ren
- Department of General Medicine, The First Affiliated Hospital of Anhui Medical University, 230000 Hefei, Anhui, China
| | - Mei Zhang
- Department of Neurology, The First Hospital of Anhui University of Science and Technology (Huainan First People's Hospital), 232000 Huainan, Anhui, China
| |
Collapse
|
3
|
Fernström E, Jarfelt M, Blomstrand M, Lannering B, Axelsson M, Wasling P, Björk‐Eriksson T, Zetterberg H, Kalm M. CSF biomarkers of neurotoxicity in childhood cancer survivors after cranial radiotherapy or surgery. Ann Clin Transl Neurol 2024; 11:2382-2391. [PMID: 39030984 PMCID: PMC11537133 DOI: 10.1002/acn3.52152] [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: 04/14/2024] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE Treatment of pediatric brain tumors is associated with potential long-term cognitive sequelae. Patients treated with craniospinal irradiation for posterior fossa tumors are at high risk. New biomarkers that could help to differentiate treatment effects from other causes of cognitive dysfunction would be valuable in tailoring optimal survivorship care. Biomarkers that reflect biological mechanisms behind treatment-associated cognitive decline would also be important in the evaluation of future treatment regimens for pediatric brain or skull base tumors. METHODS In this biomarker-finding study, 10 adult survivors of pediatric medulloblastoma, skull base tumors, and posterior fossa low-grade glioma underwent study specific lumbar puncture at a minimum of 17 years following treatment. We analyzed cerebrospinal fluid biomarkers reflecting neuron and astrocyte integrity, amyloid metabolism, inflammation, extracellular matrix, synaptic integrity, and blood-brain barrier function. The values were compared with biomarker levels in healthy controls of comparable age. RESULTS Biomarkers reflecting neuronal injury (neurofilament light chain protein), astrocyte injury or activation (glial fibrillary acidic protein) as well as inflammation (YKL-40) were significantly elevated in cancer survivors compared to controls. Biomarkers reflecting amyloid metabolism showed a pattern of decrease in patients treated for medulloblastoma. INTERPRETATION The results suggest a potential chronic low-grade neurodegeneration and astrocyte activation in patients treated for pediatric brain or skull base tumors. Protein biomarkers of CNS disease could potentially be used to increase our understanding of the contribution from different tumor treatments with regard to long-term symptoms in cancer patients.
Collapse
Affiliation(s)
- Erik Fernström
- Department of OncologyInstitute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University HospitalGothenburgSweden
| | - Marianne Jarfelt
- Department of OncologyInstitute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University HospitalGothenburgSweden
| | - Malin Blomstrand
- Department of OncologyInstitute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University HospitalGothenburgSweden
| | - Birgitta Lannering
- Department of PediatricsInstitute of Clinical Sciences, Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Markus Axelsson
- Department of Clinical NeuroscienceInstitute of Neuroscience and Physiology at the Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Pontus Wasling
- Department of Clinical NeuroscienceInstitute of Neuroscience and Physiology at the Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Thomas Björk‐Eriksson
- Department of OncologyInstitute of Clinical Sciences, Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Regional Cancer Centre WestWestern Sweden Healthcare RegionGothenburgSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong Kong, China
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public Health, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Marie Kalm
- Department of PharmacologyInstitute of Neuroscience and Physiology, Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| |
Collapse
|
4
|
Schauer SP, Cho CH, Novikova G, Roth GA, Lee J, Sharma AD, Foley AR, Ng C, Shen P, Choi M, Ma TP, Phu L, Budayeva HG, Cheung TK, Lalehzadeh G, Imperio J, Ngu H, Etxeberria A, Liang Y, Rezzonico MG, Dourado M, Huang K, Lai Z, Hokom M, Pandya NJ, Newton D, Abdel‐Haleem AM, Chan P, Lee D, Tassew NG, Sangaraju D, O'Connor D, Hötzel I, Stark KL, Chou C, Foreman O, Easton A, Wildsmith KR, Sperinde G, Rose CM, Friedman BA, Fuji RN, Weimer RM, Meilandt WJ, Sadekar S, Nugent AA, Biever A. Primate cerebrospinal fluid CHI3L1 reflects brain TREM2 agonism. Alzheimers Dement 2024; 20:5861-5888. [PMID: 39090679 PMCID: PMC11497760 DOI: 10.1002/alz.13921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Triggering receptor expressed on myeloid cells 2 (TREM2) agonists are being clinically evaluated as disease-modifying therapeutics for Alzheimer's disease. Clinically translatable pharmacodynamic (PD) biomarkers are needed to confirm drug activity and select the appropriate therapeutic dose in clinical trials. METHODS We conducted multi-omic analyses on paired non-human primate brain and cerebrospinal fluid (CSF), and stimulation of human induced pluripotent stem cell-derived microglia cultures after TREM2 agonist treatment, followed by validation of candidate fluid PD biomarkers using immunoassays. We immunostained microglia to characterize proliferation and clustering. RESULTS We report CSF soluble TREM2 (sTREM2) and CSF chitinase-3-like protein 1 (CHI3L1/YKL-40) as PD biomarkers for the TREM2 agonist hPara.09. The respective reduction of sTREM2 and elevation of CHI3L1 in brain and CSF after TREM2 agonist treatment correlated with transient microglia proliferation and clustering. DISCUSSION CSF CHI3L1 and sTREM2 reflect microglial TREM2 agonism and can be used as clinical PD biomarkers to monitor TREM2 activity in the brain. HIGHLIGHTS CSF soluble triggering receptor expressed on myeloid cells 2 (sTREM2) reflects brain target engagement for a novel TREM2 agonist, hPara.09. CSF chitinase-3-like protein 1 reflects microglial TREM2 agonism. Both can be used as clinical fluid biomarkers to monitor TREM2 activity in brain.
Collapse
Affiliation(s)
- Stephen P. Schauer
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Chang Hoon Cho
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gloriia Novikova
- Department of BioinformaticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gillie A. Roth
- Department of Preclinical and Translational Pharmacokinetics and PharmacodynamicsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Julie Lee
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Anup D. Sharma
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Alejandro R. Foley
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Carl Ng
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Philip Shen
- Department of Safety Assessment PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Meena Choi
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Taylur P. Ma
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Lilian Phu
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Hanna G. Budayeva
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Tommy K. Cheung
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Guita Lalehzadeh
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Jose Imperio
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Hai Ngu
- Department of Research PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Ainhoa Etxeberria
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Yuxin Liang
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | | | - Michelle Dourado
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kevin Huang
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Zijuan Lai
- Department of Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Martha Hokom
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Nikhil J. Pandya
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Dwight Newton
- Roche InformaticsHoffmann‐La Roche, Ltd.MississaugaOntarioCanada
| | | | - Pamela Chan
- Department of Biochemical and Cellular PharmacologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Donna Lee
- Department of Safety Assessment ToxicologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Nardos G. Tassew
- Department of Safety Assessment ToxicologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Dewakar Sangaraju
- Department of Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Deborah O'Connor
- Department of ChemistryManufacturing, and ControlsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Isidro Hötzel
- Department of Antibody EngineeringGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kimberly L. Stark
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Carolina Chou
- Department of Safety Assessment Nonclinical OperationsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Oded Foreman
- Department of Research PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Amy Easton
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kristin R. Wildsmith
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gizette Sperinde
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Christopher M. Rose
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Brad A. Friedman
- Department of BioinformaticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Reina N. Fuji
- Department of Safety Assessment PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Robby M. Weimer
- Department of Translational ImagingGenentech, Inc.South San FranciscoCaliforniaUSA
| | | | - Shraddha Sadekar
- Department of Preclinical and Translational Pharmacokinetics and PharmacodynamicsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Alicia A. Nugent
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Anne Biever
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| |
Collapse
|
5
|
Gevezova M, Ivanov Z, Pacheva I, Timova E, Kazakova M, Kovacheva E, Ivanov I, Sarafian V. Bioenergetic and Inflammatory Alterations in Regressed and Non-Regressed Patients with Autism Spectrum Disorder. Int J Mol Sci 2024; 25:8211. [PMID: 39125780 PMCID: PMC11311370 DOI: 10.3390/ijms25158211] [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: 06/20/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Autism spectrum disorder (ASD) is associated with multiple physiological abnormalities. Current laboratory and clinical evidence most commonly report mitochondrial dysfunction, oxidative stress, and immunological imbalance in almost every cell type of the body. The present work aims to evaluate oxygen consumption rate (OCR), extracellular acidification rate (ECAR), and inflammation-related molecules such as Cyclooxygenase-2 (COX-2), chitinase 3-like protein 1 (YKL-40), Interleukin-1 beta (IL-1β), Interleukin-9 (IL-9) in ASD children with and without regression compared to healthy controls. Children with ASD (n = 56) and typically developing children (TDC, n = 12) aged 1.11 to 11 years were studied. Mitochondrial activity was examined in peripheral blood mononuclear cells (PBMCs) isolated from children with ASD and from the control group, using a metabolic analyzer. Gene and protein levels of IL-1β, IL-9, COX-2, and YKL-40 were investigated in parallel. Our results showed that PBMCs of the ASD subgroup of regressed patients (ASD R(+), n = 21) had a specific pattern of mitochondrial activity with significantly increased maximal respiration, respiratory spare capacity, and proton leak compared to the non-regressed group (ASD R(-), n = 35) and TDC. Furthermore, we found an imbalance in the studied proinflammatory molecules and increased levels in ASD R(-) proving the involvement of inflammatory changes. The results of this study provide new evidence for specific bioenergetic profiles of immune cells and elevated inflammation-related molecules in ASD. For the first time, data on a unique metabolic profile in ASD R(+) and its comparison with a random group of children of similar age and sex are provided. Our data show that mitochondrial dysfunction is more significant in ASD R(+), while in ASD R(-) inflammation is more pronounced. Probably, in the group without regression, immune mechanisms (immune dysregulation, leading to inflammation) begin initially, and at a later stage mitochondrial activity is also affected under exogenous factors. On the other hand, in the regressed group, the initial damage is in the mitochondria, and perhaps at a later stage immune dysfunction is involved.
Collapse
Affiliation(s)
- Maria Gevezova
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
- Research Institute at MU-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Zdravko Ivanov
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
| | - Iliana Pacheva
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (I.P.); (I.I.)
- Pediatrics Clinic, St. George University Hospital, 4002 Plovdiv, Bulgaria;
| | - Elena Timova
- Pediatrics Clinic, St. George University Hospital, 4002 Plovdiv, Bulgaria;
| | - Maria Kazakova
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
- Research Institute at MU-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Eleonora Kovacheva
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
- Research Institute at MU-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Ivan Ivanov
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (I.P.); (I.I.)
- Pediatrics Clinic, St. George University Hospital, 4002 Plovdiv, Bulgaria;
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
- Research Institute at MU-Plovdiv, 4002 Plovdiv, Bulgaria
| |
Collapse
|
6
|
Bonanni R, Cariati I, Cifelli P, Frank C, Annino G, Tancredi V, D'Arcangelo G. Exercise to Counteract Alzheimer's Disease: What Do Fluid Biomarkers Say? Int J Mol Sci 2024; 25:6951. [PMID: 39000060 PMCID: PMC11241657 DOI: 10.3390/ijms25136951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Neurodegenerative diseases (NDs) represent an unsolved problem to date with an ever-increasing population incidence. Particularly, Alzheimer's disease (AD) is the most widespread ND characterized by an accumulation of amyloid aggregates of beta-amyloid (Aβ) and Tau proteins that lead to neuronal death and subsequent cognitive decline. Although neuroimaging techniques are needed to diagnose AD, the investigation of biomarkers within body fluids could provide important information on neurodegeneration. Indeed, as there is no definitive solution for AD, the monitoring of these biomarkers is of strategic importance as they are useful for both diagnosing AD and assessing the progression of the neurodegenerative state. In this context, exercise is known to be an effective non-pharmacological management strategy for AD that can counteract cognitive decline and neurodegeneration. However, investigation of the concentration of fluid biomarkers in AD patients undergoing exercise protocols has led to unclear and often conflicting results, suggesting the need to clarify the role of exercise in modulating fluid biomarkers in AD. Therefore, this critical literature review aims to gather evidence on the main fluid biomarkers of AD and the modulatory effects of exercise to clarify the efficacy and usefulness of this non-pharmacological strategy in counteracting neurodegeneration in AD.
Collapse
Affiliation(s)
- Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Pierangelo Cifelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Claudio Frank
- UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Giuseppe Annino
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Sports Engineering Laboratory, Department of Industrial Engineering, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Giovanna D'Arcangelo
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| |
Collapse
|
7
|
Ham HJ, Lee YS, Koo JK, Yun J, Son DJ, Han SB, Hong JT. Inhibition of Amyloid-β (Aβ)-Induced Cognitive Impairment and Neuroinflammation in CHI3L1 Knockout Mice through Downregulation of ERK-PTX3 Pathway. Int J Mol Sci 2024; 25:5550. [PMID: 38791588 PMCID: PMC11122210 DOI: 10.3390/ijms25105550] [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: 03/15/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Several clinical studies reported that the elevated expression of Chitinase-3-like 1 (CHI3L1) was observed in patients suffering from a wide range of diseases: cancer, metabolic, and neurological diseases. However, the role of CHI3L1 in AD is still unclear. Our previous study demonstrated that 2-({3-[2-(1-Cyclohexen-1-yl)ethyl]-6,7-dimethoxy-4-oxo-3,4-dihydro-2-quinazolinyl}culfanyl)-N-(4-ethylphenyl)butanamide, a CHI3L1 inhibiting compound, alleviates memory and cognitive impairment and inhibits neuroinflammation in AD mouse models. In this study, we studied the detailed correlation of CHI3L1 and AD using serum from AD patients and using CHI3L1 knockout (KO) mice with Aβ infusion (300 pmol/day, 14 days). Serum levels of CHI3L1 were significantly elevated in patients with AD compared to normal subjects, and receiver operating characteristic (ROC) analysis data based on serum analysis suggested that CHI3L1 could be a significant diagnostic reference for AD. To reveal the role of CHI3L1 in AD, we investigated the CHI3L1 deficiency effect on memory impairment in Aβ-infused mice and microglial BV-2 cells. In CHI3L1 KO mice, Aβ infusion resulted in lower levels of memory dysfunction and neuroinflammation compared to that of WT mice. CHI3L1 deficiency selectively inhibited phosphorylation of ERK and IκB as well as inhibition of neuroinflammation-related factors in vivo and in vitro. On the other hand, treatment with recombinant CHI3L1 increased neuroinflammation-related factors and promoted phosphorylation of IκB except for ERK in vitro. Web-based gene network analysis and our results showed that CHI3L1 is closely correlated with PTX3. Moreover, in AD patients, we found that serum levels of PTX3 were correlated with serum levels of CHI3L1 by Spearman correlation analysis. These results suggest that CHI3L1 deficiency could inhibit AD development by blocking the ERK-dependent PTX3 pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju 28160, Republic of Korea
| |
Collapse
|
8
|
Czestkowski W, Krzemiński Ł, Piotrowicz MC, Mazur M, Pluta E, Andryianau G, Koralewski R, Matyszewski K, Olejniczak S, Kowalski M, Lisiecka K, Kozieł R, Piwowar K, Papiernik D, Nowotny M, Napiórkowska-Gromadzka A, Nowak E, Niedziałek D, Wieczorek G, Siwińska A, Rejczak T, Jędrzejczak K, Mulewski K, Olczak J, Zasłona Z, Gołębiowski A, Drzewicka K, Bartoszewicz A. Structure-Based Discovery of High-Affinity Small Molecule Ligands and Development of Tool Probes to Study the Role of Chitinase-3-Like Protein 1. J Med Chem 2024; 67:3959-3985. [PMID: 38427954 DOI: 10.1021/acs.jmedchem.3c02255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Chitinase-3-like-1 (CHI3L1), also known as YKL-40, is a glycoprotein linked to inflammation, fibrosis, and cancer. This study explored CHI3L1's interactions with various oligosaccharides using microscale thermophoresis (MST) and AlphaScreen (AS). These investigations guided the development of high-throughput screening assays to assess interference of small molecules in binding between CHI3L1 and biotinylated small molecules or heparan sulfate-based probes. Small molecule binders of YKL-40 were identified in our chitotriosidase inhibitors library with MST and confirmed through X-ray crystallography. Based on cocrystal structures of potent hit compounds with CHI3L1, small molecule probes 19 and 20 were designed for an AS assay. Structure-based optimization led to compounds 30 and 31 with nanomolar activities and drug-like properties. Additionally, an orthogonal AS assay using biotinylated heparan sulfate as a probe was developed. The compounds' affinity showed a significant correlation in both assays. These screening tools and compounds offer novel avenues for investigating the role of CHI3L1.
Collapse
Affiliation(s)
| | | | | | - Marzena Mazur
- Molecure S.A., Żwirki I Wigury 101, Warsaw 02-089, Poland
| | - Elżbieta Pluta
- Molecure S.A., Żwirki I Wigury 101, Warsaw 02-089, Poland
| | | | | | | | | | | | | | - Rafał Kozieł
- Molecure S.A., Żwirki I Wigury 101, Warsaw 02-089, Poland
| | | | | | - Marcin Nowotny
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, Warsaw 02-109, Poland
| | - Agnieszka Napiórkowska-Gromadzka
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, Warsaw 02-109, Poland
| | - Elżbieta Nowak
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology in Warsaw, Ks. Trojdena 4, Warsaw 02-109, Poland
| | | | | | - Anna Siwińska
- Molecure S.A., Żwirki I Wigury 101, Warsaw 02-089, Poland
| | - Tomasz Rejczak
- Molecure S.A., Żwirki I Wigury 101, Warsaw 02-089, Poland
| | | | | | - Jacek Olczak
- Molecure S.A., Żwirki I Wigury 101, Warsaw 02-089, Poland
| | | | | | | | | |
Collapse
|
9
|
Pase MP, Himali JJ, Puerta R, Beiser AS, Gonzales MM, Satizabal CL, Yang Q, Aparicio HJ, Kojis DJ, Decarli CS, Lopez OL, Longstreth W, Gudnason V, Mosley TH, Bis JC, Fohner A, Psaty BM, Boada M, García-González P, Valero S, Marquié M, Tracy R, Launer LJ, Ruiz A, Fornage M, Seshadri S. Association of Plasma YKL-40 With MRI, CSF, and Cognitive Markers of Brain Health and Dementia. Neurology 2024; 102:e208075. [PMID: 38290090 PMCID: PMC11383876 DOI: 10.1212/wnl.0000000000208075] [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: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Higher YKL-40 levels in the CSF are a known biomarker of brain inflammation. We explored the utility of plasma YKL-40 as a biomarker for accelerated brain aging and dementia risk. METHODS We performed cross-sectional and prospective analyses of 4 community-based cohorts in the United States or Europe: the Age, Gene/Environment Susceptibility-Reykjavik Study, Atherosclerosis Risk in the Communities study, Coronary Artery Risk Development in Young Adults study, and Framingham Heart Study (FHS). YKL-40 was measured from stored plasma by a single laboratory using Mesoscale Discovery with levels log transformed and standardized within each cohort. Outcomes included MRI total brain volume, hippocampal volume, and white matter hyperintensity volume (WMHV) as a percentage of intracranial volume, a general cognitive composite derived from neuropsychological testing (SD units [SDU]), and the risk of incident dementia. We sought to replicate associations with dementia in the clinic-based ACE csf cohort, which also had YKL-40 measured from the CSF. RESULTS Meta-analyses of MRI outcomes included 6,558 dementia-free participants, and for analysis of cognition, 6,670. The blood draw preceded MRI/cognitive assessment by up to 10.6 years across cohorts. The mean ages ranged from 50 to 76 years, with 39%-48% male individuals. In random-effects meta-analysis of study estimates, each SDU increase in log-transformed YKL-40 levels was associated with smaller total brain volume (β = -0.33; 95% CI -0.45 to -0.22; p < 0.0001) and poorer cognition (β = -0.04; 95% CI -0.07 to -0.02; p < 0.01), following adjustments for demographic variables. YKL-40 levels did not associate with hippocampal volume or WMHV. In the FHS, each SDU increase in log YKL-40 levels was associated with a 64% increase in incident dementia risk over a median of 5.8 years of follow-up, following adjustments for demographic variables (hazard ratio 1.64; 95% CI 1.25-2.16; p < 0.001). In the ACE csf cohort, plasma and CSF YKL-40 were correlated (r = 0.31), and both were associated with conversion from mild cognitive impairment to dementia, independent of amyloid, tau, and neurodegeneration status. DISCUSSION Higher plasma YKL-40 levels were associated with lower brain volume, poorer cognition, and incident dementia. Plasma YKL-40 may be useful for studying the association of inflammation and its treatment on dementia risk.
Collapse
Affiliation(s)
- Matthew P Pase
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Jayandra J Himali
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Raquel Puerta
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Alexa S Beiser
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Mitzi M Gonzales
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Claudia L Satizabal
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Qiong Yang
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Hugo J Aparicio
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Daniel J Kojis
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Charles S Decarli
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Oscar L Lopez
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Will Longstreth
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Vilmundur Gudnason
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Thomas H Mosley
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Joshua C Bis
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Alison Fohner
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Bruce M Psaty
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Mercè Boada
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Pablo García-González
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Sergi Valero
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Marta Marquié
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Russell Tracy
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Lenore J Launer
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Agustín Ruiz
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Myriam Fornage
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Sudha Seshadri
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| |
Collapse
|
10
|
Yu JE, Yeo IJ, Han SB, Yun J, Kim B, Yong YJ, Lim YS, Kim TH, Son DJ, Hong JT. Significance of chitinase-3-like protein 1 in the pathogenesis of inflammatory diseases and cancer. Exp Mol Med 2024; 56:1-18. [PMID: 38177294 PMCID: PMC10834487 DOI: 10.1038/s12276-023-01131-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/06/2023] [Accepted: 08/28/2023] [Indexed: 01/06/2024] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that mediates inflammation, macrophage polarization, apoptosis, and carcinogenesis. The expression of CHI3L1 is strongly upregulated by various inflammatory and immunological diseases, including several cancers, Alzheimer's disease, and atherosclerosis. Several studies have shown that CHI3L1 can be considered as a marker of disease diagnosis, prognosis, disease activity, and severity. In addition, the proinflammatory action of CHI3L1 may be mediated via responses to various proinflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, interleukin-6, and interferon-γ. Therefore, CHI3L1 may contribute to a vast array of inflammatory diseases. However, its pathophysiological and pharmacological roles in the development of inflammatory diseases remain unclear. In this article, we review recent findings regarding the roles of CHI3L1 in the development of inflammatory diseases and suggest therapeutic approaches that target CHI3L1.
Collapse
Affiliation(s)
- Ji Eun Yu
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Bongcheol Kim
- Senelix Co. Ltd., 25, Beobwon-ro 11-gil, Songpa-gu, Seoul, 05836, Republic of Korea
| | - Yoon Ji Yong
- PRESTI GEBIOLOGICS Co. Ltd., Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28161, Republic of Korea
| | - Young-Soo Lim
- PRESTI GEBIOLOGICS Co. Ltd., Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28161, Republic of Korea
| | - Tae Hun Kim
- Autotelic Bio Inc., Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
11
|
Selahaddin E, Bahadir D, Sengul S, Seyithan T, Hasan U, Gülcin E, Abdurrahman A. Serum Galectin-1, Galectin-9, and YKL-40 levels in bipolar disorder and their relationship with cognitive functions. Brain Behav 2024; 14:e3421. [PMID: 38346720 PMCID: PMC10861353 DOI: 10.1002/brb3.3421] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 01/16/2024] [Accepted: 01/21/2024] [Indexed: 02/15/2024] Open
Abstract
PURPOSE The number of studies conducted on the role of neuroinflammation in the etiopathogenesis of bipolar disorder has been increasing in recent years. The role of Galectin-1, Galectin-9, and YKL-40, which are considered to play roles in neuroinflammation and the etiopathogenesis of bipolar disorder, and the relationship of these parameters with cognitive functions were investigated in the present study. METHOD Serum Galectin-1, Galectin-9, and YKL-40 levels were measured with the ELISA Method in 64 bipolar euthymic patients and 64 healthy controls. The Stroop and trail-making tests were administered to assess cognitive functions in all participants. RESULTS Serum Galectin-1, Galectin-9, and YKL-40 levels were statistically and significantly lower in the patient group when compared to the healthy control group. The scores of the Stroop test and trail-making tests were statistically higher in the patient group than in the healthy control group. There was a weak and positive correlation between serum Galectin-1, Galectin-9, and YKL-40 levels and cognitive performance in all participants. DISCUSSION AND CONCLUSION Statistically significant low levels of serum Galectin-1, Galectin-9, and YKL-40 detected in the patient group suggest that these parameters have important roles in neuroinflammation. The statistically higher Stroop and trail-making test scores of the patient group compared to the control group indicates that the cognitive performance of the patient group was weaker. Also, the positive correlation between Galectin-1, Galectin-9, and YKL-40 levels and cognitive performance suggests that these molecules may have a neuroprotective role. We think that the present study will contribute to this field where there is very limited data in the literature.
Collapse
Affiliation(s)
- Elcicek Selahaddin
- Faculty of MedicineDepartment of PsychiatryGaziantep UniversityGaziantepTurkey
| | - Demir Bahadir
- Faculty of MedicineDepartment of PsychiatryGaziantep UniversityGaziantepTurkey
| | - Sahin Sengul
- Faculty of MedicineDepartment of PsychiatryGaziantep UniversityGaziantepTurkey
| | - Taysi Seyithan
- Faculty of MedicineDepartment of BiochemistryGaziantep UniversityGaziantepTurkey
| | - Ulusal Hasan
- Faculty of MedicineDepartment of BiochemistryGaziantep UniversityGaziantepTurkey
| | - Elboga Gülcin
- Faculty of MedicineDepartment of PsychiatryGaziantep UniversityGaziantepTurkey
| | | |
Collapse
|
12
|
Kong F, Wu T, Dai J, Cai J, Zhai Z, Zhu Z, Xu Y, Sun T. Knowledge domains and emerging trends of Genome-wide association studies in Alzheimer's disease: A bibliometric analysis and visualization study from 2002 to 2022. PLoS One 2024; 19:e0295008. [PMID: 38241287 PMCID: PMC10798548 DOI: 10.1371/journal.pone.0295008] [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: 04/20/2023] [Accepted: 11/13/2023] [Indexed: 01/21/2024] Open
Abstract
OBJECTIVES Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive decline in cognitive and behavioral function. Studies have shown that genetic factors are one of the main causes of AD risk. genome-wide association study (GWAS), as a novel and effective tool for studying the genetic risk of diseases, has attracted attention from researchers in recent years and a large number of studies have been conducted. This study aims to summarize the literature on GWAS in AD by bibliometric methods, analyze the current status, research hotspots and future trends in this field. METHODS We retrieved articles on GWAS in AD published between 2002 and 2022 from Web of Science. CiteSpace and VOSviewer software were applied to analyze the articles for the number of articles published, countries/regions and institutions of publication, authors and cited authors, highly cited literature, and research hotspots. RESULTS We retrieved a total of 2,751 articles. The United States had the highest number of publications in this field, and Columbia University was the institution with the most published articles. The identification of AD-related susceptibility genes and their effects on AD is one of the current research hotspots. Numerous risk genes have been identified, among which APOE, CLU, CD2AP, CD33, EPHA1, PICALM, CR1, ABCA7 and TREM2 are the current genes of interest. In addition, risk prediction for AD and research on other related diseases are also popular research directions in this field. CONCLUSION This study conducted a comprehensive analysis of GWAS in AD and identified the current research hotspots and research trends. In addition, we also pointed out the shortcomings of current research and suggested future research directions. This study can provide researchers with information about the knowledge structure and emerging trends in the field of GWAS in AD and provide guidance for future research.
Collapse
Affiliation(s)
- Fanjing Kong
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianyu Wu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingyi Dai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Cai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenwei Zhai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhishan Zhu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Sun
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Bhalala OG, Watson R, Yassi N. Multi-Omic Blood Biomarkers as Dynamic Risk Predictors in Late-Onset Alzheimer's Disease. Int J Mol Sci 2024; 25:1231. [PMID: 38279230 PMCID: PMC10816901 DOI: 10.3390/ijms25021231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Late-onset Alzheimer's disease is the leading cause of dementia worldwide, accounting for a growing burden of morbidity and mortality. Diagnosing Alzheimer's disease before symptoms are established is clinically challenging, but would provide therapeutic windows for disease-modifying interventions. Blood biomarkers, including genetics, proteins and metabolites, are emerging as powerful predictors of Alzheimer's disease at various timepoints within the disease course, including at the preclinical stage. In this review, we discuss recent advances in such blood biomarkers for determining disease risk. We highlight how leveraging polygenic risk scores, based on genome-wide association studies, can help stratify individuals along their risk profile. We summarize studies analyzing protein biomarkers, as well as report on recent proteomic- and metabolomic-based prediction models. Finally, we discuss how a combination of multi-omic blood biomarkers can potentially be used in memory clinics for diagnosis and to assess the dynamic risk an individual has for developing Alzheimer's disease dementia.
Collapse
Affiliation(s)
- Oneil G. Bhalala
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| | - Rosie Watson
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| | - Nawaf Yassi
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; (R.W.); (N.Y.)
- Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3050, Australia
| |
Collapse
|
14
|
Gaikwad S, Senapati S, Haque MA, Kayed R. Senescence, brain inflammation, and oligomeric tau drive cognitive decline in Alzheimer's disease: Evidence from clinical and preclinical studies. Alzheimers Dement 2024; 20:709-727. [PMID: 37814508 PMCID: PMC10841264 DOI: 10.1002/alz.13490] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/11/2023]
Abstract
Aging, tau pathology, and chronic inflammation in the brain play crucial roles in synaptic loss, neurodegeneration, and cognitive decline in tauopathies, including Alzheimer's disease. Senescent cells accumulate in the aging brain, accelerate the aging process, and promote tauopathy progression through their abnormal inflammatory secretome known as the senescence-associated secretory phenotype (SASP). Tau oligomers (TauO)-the most neurotoxic tau species-are known to induce senescence and the SASP, which subsequently promote neuropathology, inflammation, oxidative stress, synaptic dysfunction, neuronal death, and cognitive dysfunction. TauO, brain inflammation, and senescence are associated with heterogeneity in tauopathy progression and cognitive decline. However, the underlying mechanisms driving the disease heterogeneity remain largely unknown, impeding the development of therapies for tauopathies. Based on clinical and preclinical evidence, this review highlights the critical role of TauO and senescence in neurodegeneration. We discuss key knowledge gaps and potential strategies for targeting senescence and TauO to treat tauopathies. HIGHLIGHTS: Senescence, oligomeric Tau (TauO), and brain inflammation accelerate the aging process and promote the progression of tauopathies, including Alzheimer's disease. We discuss their role in contributing to heterogeneity in tauopathy and cognitive decline. We highlight strategies to target senescence and TauO to treat tauopathies while addressing key knowledge gaps.
Collapse
Affiliation(s)
- Sagar Gaikwad
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Sudipta Senapati
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Md. Anzarul Haque
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
15
|
Zheng Y, Zhang X, Zhang R, Wang Z, Gan J, Gao Q, Yang L, Xu P, Jiang X. Inflammatory signaling pathways in the treatment of Alzheimer's disease with inhibitors, natural products and metabolites (Review). Int J Mol Med 2023; 52:111. [PMID: 37800614 PMCID: PMC10558228 DOI: 10.3892/ijmm.2023.5314] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023] Open
Abstract
The intricate nature of Alzheimer's disease (AD) pathogenesis poses a persistent obstacle to drug development. In recent times, neuroinflammation has emerged as a crucial pathogenic mechanism of AD, and the targeting of inflammation has become a viable approach for the prevention and management of AD. The present study conducted a comprehensive review of the literature between October 2012 and October 2022, identifying a total of 96 references, encompassing 91 distinct pharmaceuticals that have been investigated for their potential impact on AD by inhibiting neuroinflammation. Research has shown that pharmaceuticals have the potential to ameliorate AD by reducing neuroinflammation mainly through regulating inflammatory signaling pathways such as NF‑κB, MAPK, NLRP3, PPARs, STAT3, CREB, PI3K/Akt, Nrf2 and their respective signaling pathways. Among them, tanshinone IIA has been extensively studied for its anti‑inflammatory effects, which have shown significant pharmacological properties and can be applied clinically. Thus, it may hold promise as an effective drug for the treatment of AD. The present review elucidated the inflammatory signaling pathways of pharmaceuticals that have been investigated for their therapeutic efficacy in AD and elucidates their underlying mechanisms. This underscores the auspicious potential of pharmaceuticals in ameliorating AD by impeding neuroinflammation.
Collapse
Affiliation(s)
| | | | - Ruifeng Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Ziyu Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Jiali Gan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Qing Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Lin Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Pengjuan Xu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Xijuan Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| |
Collapse
|
16
|
Dubois B, von Arnim CAF, Burnie N, Bozeat S, Cummings J. Biomarkers in Alzheimer's disease: role in early and differential diagnosis and recognition of atypical variants. Alzheimers Res Ther 2023; 15:175. [PMID: 37833762 PMCID: PMC10571241 DOI: 10.1186/s13195-023-01314-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Development of in vivo biomarkers has shifted the diagnosis of Alzheimer's disease (AD) from the later dementia stages of disease towards the earlier stages and has introduced the potential for pre-symptomatic diagnosis. The International Working Group recommends that AD diagnosis is restricted in the clinical setting to people with specific AD phenotypes and supportive biomarker findings. MAIN BODY In this review, we discuss the phenotypic presentation and use of biomarkers for the early diagnosis of typical and atypical AD and describe how this can support clinical decision making, benefit patient communication, and improve the patient journey. Early diagnosis is essential to optimize the benefits of available and emerging treatments. As atypical presentations of AD often mimic other dementias, differential diagnosis can be challenging and can be facilitated using AD biomarkers. However, AD biomarkers alone are not sufficient to confidently diagnose AD or predict disease progression and should be supplementary to clinical assessment to help inform the diagnosis of AD. CONCLUSIONS Use of AD biomarkers with incorporation of atypical AD phenotypes into diagnostic criteria will allow earlier diagnosis of patients with atypical clinical presentations that otherwise would have been misdiagnosed and treated inappropriately. Early diagnosis is essential to guide informed discussion, appropriate care and support, and individualized treatment. It is hoped that disease-modifying treatments will impact the underlying AD pathology; thus, determining the patient's AD phenotype will be a critical factor in guiding the therapeutic approach and the assessment of the effects of interventions.
Collapse
Affiliation(s)
- Bruno Dubois
- Assistance Publique-Hôpitaux de Paris (AP-HP), Memory and Alzheimer's Disease Institute, Sorbonne University, Paris, France
- Brain Institute, Sorbonne University, Paris, France
| | | | - Nerida Burnie
- General Practice, South West London CCG, London, UK
- London Dementia Clinical Network, London, UK
| | | | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
17
|
Jiang W, Zhu F, Xu H, Xu L, Li H, Yang X, Khan Afridi S, Lai S, Qiu X, Liu C, Li H, Long Y, Wang Y, Connolly K, Elias JA, Lee CG, Cui Y, Huang YWA, Qiu W, Tang C. CHI3L1 signaling impairs hippocampal neurogenesis and cognitive function in autoimmune-mediated neuroinflammation. SCIENCE ADVANCES 2023; 9:eadg8148. [PMID: 37756391 PMCID: PMC10530095 DOI: 10.1126/sciadv.adg8148] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
Chitinase-3-like protein 1 (CHI3L1) is primarily secreted by activated astrocytes in the brain and is known as a reliable biomarker for inflammatory central nervous system (CNS) conditions such as neurodegeneration and autoimmune disorders like neuromyelitis optica (NMO). NMO is an astrocyte disease caused by autoantibodies targeting the astroglial protein aquaporin 4 (AQP4) and leads to vision loss, motor deficits, and cognitive decline. In this study examining CHI3L1's biological function in neuroinflammation, we found that CHI3L1 expression correlates with cognitive impairment in our NMO patient cohort. Activated astrocytes secrete CHI3L1 in response to AQP4 autoantibodies, and this inhibits the proliferation and neuronal differentiation of neural stem cells. Mouse models showed decreased hippocampal neurogenesis and impaired learning behaviors, which could be rescued by depleting CHI3L1 in astrocytes. The molecular mechanism involves CHI3L1 engaging the CRTH2 receptor and dampening β-catenin signaling for neurogenesis. Blocking this CHI3L1/CRTH2/β-catenin cascade restores neurogenesis and improves cognitive deficits, suggesting the potential for therapeutic development in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Fan Zhu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Huiming Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Li Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Haoyang Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Xin Yang
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Shabbir Khan Afridi
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Shuiqing Lai
- Department of Endocrinology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, Guangdong Province 510080, China
| | - Xiusheng Qiu
- Vaccine Research Institute, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Chunxin Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Huilu Li
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, Guangdong Province 510260, China
| | - Youming Long
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, Guangdong Province 510260, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Kevin Connolly
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Jack A. Elias
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Chun Geun Lee
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Yaxiong Cui
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Advanced Innovation Center for Structural Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yu-Wen Alvin Huang
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| | - Changyong Tang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, Guangdong Province 510630, China
| |
Collapse
|
18
|
Ma J, Huang L, Gao YB, Li MX, Chen LL, Yang L. Circ_TNFRSF21 promotes cSCC metastasis and M2 macrophage polarization via miR-214-3p/CHI3L1. J Dermatol Sci 2023; 111:32-42. [PMID: 37442735 DOI: 10.1016/j.jdermsci.2023.06.001] [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: 11/06/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (cSCC) is a highly invasive disease with the potential to metastasize and cause fatality. Therefore, it is crucial to understand the mechanism behind cSCC in order to devise effective strategies to combat this disease. OBJECTIVE We investigated the function of circ_TNFRSF21/miR-214-3p/CHI3L1 axis in cSCC. METHODS The features of circ_TNFRSF21 was characterized using Sanger sequencing, and RNase R/actinomycin D treatment. Genes and M1/M2 markers levels were assessed by qRT-PCR and IHC. The proliferation, migration, and invasion of cells were evaluated by CCK-8, colony formation, EdU incorporation, and transwell assays. Tumor growth and metastasis in vivo were evaluated by nude mouse xenograft model. Interactions of circ_TNFRSF21/miR-214-3p and miR-214-3p/CHI3L1 were validated by RNA immunoprecipitation and dual luciferase assay. RESULTS Circ_TNFRSF21 and CHI3L1 expression were elevated in both human cSCC tissues and cells, whereas miR-214-3p was reduced. Circ_TNFRSF21 silencing or miR-214-3p overexpression suppressed cSCC cell proliferation, migration, invasion, and M2 macrophage polarization. Circ_TNFRSF21 functioned as a sponge for miR-214-3p while miR-214-3p directly targeted CHI3L1. Knockdown of miR-214-3p reversed the effects of circ_TNFRSF21 knockdown on cSCC development, while CHI3L1 upregulation reversed the effects of miR-214-3p overexpression. Furthermore, knockdown of circ_TNFRSF21 inhibited cSCC tumor growth and metastasis in vivo. CONCLUSION Circ_TNFRSF21 plays a significant role in cSCC progression by enhancing cell proliferation, migration, invasion, and M2 macrophage polarization through inhibiting miR-214-3p and subsequent disinhibition of CHI3L1. These findings deepen our understanding of the molecular mechanism of cSCC and propose the circ_TNFRSF21/miR-214-3p/CHI3L1 axis as promising diagnosis markers or therapeutic targets for cSCC.
Collapse
Affiliation(s)
- Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangdong Province, PR China
| | - Lei Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangdong Province, PR China
| | - Yan-Bin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangdong Province, PR China
| | - Min-Xiong Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangdong Province, PR China
| | - Liang-Long Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangdong Province, PR China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangdong Province, PR China.
| |
Collapse
|
19
|
Beltran-Lobo P, Reid MJ, Jimenez-Sanchez M, Verkhratsky A, Perez-Nievas BG, Noble W. Astrocyte adaptation in Alzheimer's disease: a focus on astrocytic P2X7R. Essays Biochem 2023; 67:119-130. [PMID: 36449279 PMCID: PMC10011405 DOI: 10.1042/ebc20220079] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 12/02/2022]
Abstract
Astrocytes are key homeostatic and defensive cells of the central nervous system (CNS). They undertake numerous functions during development and in adulthood to support and protect the brain through finely regulated communication with other cellular elements of the nervous tissue. In Alzheimer's disease (AD), astrocytes undergo heterogeneous morphological, molecular and functional alterations represented by reactive remodelling, asthenia and loss of function. Reactive astrocytes closely associate with amyloid β (Aβ) plaques and neurofibrillary tangles in advanced AD. The specific contribution of astrocytes to AD could potentially evolve along the disease process and includes alterations in their signalling, interactions with pathological protein aggregates, metabolic and synaptic impairments. In this review, we focus on the purinergic receptor, P2X7R, and discuss the evidence that P2X7R activation contributes to altered astrocyte functions in AD. Expression of P2X7R is increased in AD brain relative to non-demented controls, and animal studies have shown that P2X7R antagonism improves cognitive and synaptic impairments in models of amyloidosis and tauopathy. While P2X7R activation can induce inflammatory signalling pathways, particularly in microglia, we focus here specifically on the contributions of astrocytic P2X7R to synaptic changes and protein aggregate clearance in AD, highlighting cell-specific roles of this purinoceptor activation that could be targeted to slow disease progression.
Collapse
Affiliation(s)
- Paula Beltran-Lobo
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, 5 Cutcombe Road, London, SE5 9RX, U.K
| | - Matthew J Reid
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, 5 Cutcombe Road, London, SE5 9RX, U.K
| | - Maria Jimenez-Sanchez
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, 5 Cutcombe Road, London, SE5 9RX, U.K
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, U.K
- Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania
| | - Beatriz G Perez-Nievas
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, 5 Cutcombe Road, London, SE5 9RX, U.K
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, 5 Cutcombe Road, London, SE5 9RX, U.K
| |
Collapse
|
20
|
Identification of hub proteins in cerebrospinal fluid as potential biomarkers of Alzheimer's disease by integrated bioinformatics. J Neurol 2023; 270:1487-1500. [PMID: 36396814 DOI: 10.1007/s00415-022-11476-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a heterogeneous neurodegenerative disease with complex pathophysiology. Therefore, the identification of novel effective fluid biomarkers is essential for Alzheimer's disease diagnosis and drug development. This study aimed to identify potential candidate hub proteins in cerebrospinal fluid for precise Alzheimer's disease diagnosis using bioinformatics methods. METHODS A total of 29 co-significant differentially expressed proteins were identified by differential protein expression analysis in four different cohorts. Functional enrichment analysis revealed that most of these proteins were enriched in pathways related to glycometabolism. Using the Least Absolute Shrinkage and Selection Operator (LASSO) and random forest feature selection methods, six hub proteins [14-3-3 protein zeta/delta (YWHAZ), SPARC-related modular calcium-binding protein 1 (SMOC1), aldolase A (ALDOA), pyruvate kinase isoenzyme type M2 (PKM), chitinase-3-like protein 1 (CHI3L1), and secreted phosphoprotein 1 (SPP1)] were identified. RESULTS These six hub proteins were upregulated in the cerebrospinal fluid of patients with Alzheimer's disease compared with cognitively unimpaired control individuals. Meanwhile, SMOC1, ALDOA, and PKM were specifically upregulated in the cerebrospinal fluid of patients with Alzheimer's disease but not in other neurodegenerative diseases. Build AD diagnostic models showed that a single hub protein or six hub proteins combination had an excellent ability to discriminate Alzheimer's disease. CONCLUSIONS In conclusion, our study suggests that these identified hub proteins, which are related to glycometabolism, may be potential biomarkers for further basic and clinical research in Alzheimer's disease.
Collapse
|
21
|
Brosseron F, Maass A, Kleineidam L, Ravichandran KA, Kolbe CC, Wolfsgruber S, Santarelli F, Häsler LM, McManus R, Ising C, Röske S, Peters O, Cosma NC, Schneider LS, Wang X, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Schott BH, Buerger K, Janowitz D, Dichgans M, Perneczky R, Rauchmann BS, Teipel S, Kilimann I, Görß D, Laske C, Munk MH, Düzel E, Yakupow R, Dobisch L, Metzger CD, Glanz W, Ewers M, Dechent P, Haynes JD, Scheffler K, Roy N, Rostamzadeh A, Spottke A, Ramirez A, Mengel D, Synofzik M, Jucker M, Latz E, Jessen F, Wagner M, Heneka MT. Serum IL-6, sAXL, and YKL-40 as systemic correlates of reduced brain structure and function in Alzheimer's disease: results from the DELCODE study. Alzheimers Res Ther 2023; 15:13. [PMID: 36631909 PMCID: PMC9835320 DOI: 10.1186/s13195-022-01118-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/06/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Neuroinflammation constitutes a pathological hallmark of Alzheimer's disease (AD). Still, it remains unresolved if peripheral inflammatory markers can be utilized for research purposes similar to blood-based beta-amyloid and neurodegeneration measures. We investigated experimental inflammation markers in serum and analyzed interrelations towards AD pathology features in a cohort with a focus on at-risk stages of AD. METHODS Data of 74 healthy controls (HC), 99 subjective cognitive decline (SCD), 75 mild cognitive impairment (MCI), 23 AD relatives, and 38 AD subjects were obtained from the DELCODE cohort. A panel of 20 serum biomarkers was determined using immunoassays. Analyses were adjusted for age, sex, APOE status, and body mass index and included correlations between serum and CSF marker levels and AD biomarker levels. Group-wise comparisons were based on screening diagnosis and routine AD biomarker-based schematics. Structural imaging data were combined into composite scores representing Braak stage regions and related to serum biomarker levels. The Preclinical Alzheimer's Cognitive Composite (PACC5) score was used to test for associations between the biomarkers and cognitive performance. RESULTS Each experimental marker displayed an individual profile of interrelations to AD biomarkers, imaging, or cognition features. Serum-soluble AXL (sAXL), IL-6, and YKL-40 showed the most striking associations. Soluble AXL was significantly elevated in AD subjects with pathological CSF beta-amyloid/tau profile and negatively related to structural imaging and cognitive function. Serum IL-6 was negatively correlated to structural measures of Braak regions, without associations to corresponding IL-6 CSF levels or other AD features. Serum YKL-40 correlated most consistently to CSF AD biomarker profiles and showed the strongest negative relations to structure, but none to cognitive outcomes. CONCLUSIONS Serum sAXL, IL-6, and YKL-40 relate to different AD features, including the degree of neuropathology and cognitive functioning. This may suggest that peripheral blood signatures correspond to specific stages of the disease. As serum markers did not reflect the corresponding CSF protein levels, our data highlight the need to interpret serum inflammatory markers depending on the respective protein's specific biology and cellular origin. These marker-specific differences will have to be considered to further define and interpret blood-based inflammatory profiles for AD research.
Collapse
Affiliation(s)
- Frederic Brosseron
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Anne Maass
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Luca Kleineidam
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Kishore Aravind Ravichandran
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Carl-Christian Kolbe
- grid.15090.3d0000 0000 8786 803XInstitute of Innate Immunity, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany ,grid.420044.60000 0004 0374 4101Bayer AG, Alfred-Nobel-Straße 50, 40789 Monheim am Rhein, Germany
| | - Steffen Wolfsgruber
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Francesco Santarelli
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Lisa M. Häsler
- grid.10392.390000 0001 2190 1447Hertie Institute for Clinical Brain Research, Department Cellular Neurology, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany
| | - Róisín McManus
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christina Ising
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany ,grid.452408.fExcellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany
| | - Sandra Röske
- grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Oliver Peters
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicoleta-Carmen Cosma
- grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Luisa-Sophie Schneider
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Xiao Wang
- grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Josef Priller
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany ,grid.6936.a0000000123222966Department of Psychiatry and Psychotherapy, Technical University Munich, 81675 Munich, Germany
| | - Eike J. Spruth
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Slawek Altenstein
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Anja Schneider
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Klaus Fliessbach
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jens Wiltfang
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Göttingen, Germany ,grid.7450.60000 0001 2364 4210Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany ,grid.7311.40000000123236065Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Björn H. Schott
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Göttingen, Germany ,grid.7450.60000 0001 2364 4210Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany ,grid.418723.b0000 0001 2109 6265Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Katharina Buerger
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany ,grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Daniel Janowitz
- grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Martin Dichgans
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany ,grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Robert Perneczky
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany ,grid.411095.80000 0004 0477 2585Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany ,grid.7445.20000 0001 2113 8111Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK ,grid.11835.3e0000 0004 1936 9262Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Boris-Stephan Rauchmann
- grid.411095.80000 0004 0477 2585Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Teipel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Str. 20, 18147 Rostock, Germany ,grid.413108.f0000 0000 9737 0454Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Ingo Kilimann
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Str. 20, 18147 Rostock, Germany ,grid.413108.f0000 0000 9737 0454Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Doreen Görß
- grid.413108.f0000 0000 9737 0454Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Christoph Laske
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany ,grid.10392.390000 0001 2190 1447Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Matthias H. Munk
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany ,grid.10392.390000 0001 2190 1447Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Emrah Düzel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany ,grid.5807.a0000 0001 1018 4307Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Renat Yakupow
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Laura Dobisch
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Coraline D. Metzger
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany ,grid.5807.a0000 0001 1018 4307Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany ,grid.5807.a0000 0001 1018 4307Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany
| | - Wenzel Glanz
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Michael Ewers
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Peter Dechent
- grid.7450.60000 0001 2364 4210MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University, Goettingen, Germany
| | - John Dylan Haynes
- grid.6363.00000 0001 2218 4662Bernstein Center for Computational Neurosciences, Charité – Universitätsmedizin, Berlin, Germany
| | - Klaus Scheffler
- grid.10392.390000 0001 2190 1447Department for Biomedical Magnetic Resonance, University of Tübingen, 72076 Tübingen, Germany
| | - Nina Roy
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Ayda Rostamzadeh
- grid.6190.e0000 0000 8580 3777Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Annika Spottke
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.10388.320000 0001 2240 3300Department of Neurology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alfredo Ramirez
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany ,grid.452408.fExcellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany ,grid.6190.e0000 0000 8580 3777Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany ,Department of Psychiatry & Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX USA
| | - David Mengel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany ,grid.10392.390000 0001 2190 1447Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Matthis Synofzik
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany ,grid.10392.390000 0001 2190 1447Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Mathias Jucker
- grid.10392.390000 0001 2190 1447Hertie Institute for Clinical Brain Research, Department Cellular Neurology, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany
| | - Eicke Latz
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XInstitute of Innate Immunity, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Frank Jessen
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.452408.fExcellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany ,grid.6190.e0000 0000 8580 3777Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Michael Wagner
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Michael T. Heneka
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany ,grid.16008.3f0000 0001 2295 9843Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, 4362 Esch-sur- Alzette, Luxembourg
| | | |
Collapse
|
22
|
Connolly K, Lehoux M, O’Rourke R, Assetta B, Erdemir GA, Elias JA, Lee CG, Huang YWA. Potential role of chitinase-3-like protein 1 (CHI3L1/YKL-40) in neurodegeneration and Alzheimer's disease. Alzheimers Dement 2023; 19:9-24. [PMID: 35234337 PMCID: PMC9437141 DOI: 10.1002/alz.12612] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 01/18/2023]
Abstract
Chitinase-3-like protein 1 (CHI3L1/YKL-40) has long been known as a biomarker for early detection of neuroinflammation and disease diagnosis of Alzheimer's disease (AD). In the brain, CHI3L1 is primarily provided by astrocytes and heralds the reactive, neurotoxic state triggered by inflammation and other stress signals. However, how CHI3L1 acts in neuroinflammation or how it contributes to AD and relevant neurodegenerative conditions remains unknown. In peripheral tissues, our group and others have uncovered that CHI3L1 is a master regulator for a wide range of injury and repair events, including the innate immunity pathway that resembles the neuroinflammation process governed by microglia and astrocytes. Based on assessment of current knowledge regarding CHI3L1 biology, we hypothesize that CHI3L1 functions as a signaling molecule mediating distinct neuroinflammatory responses in brain cells and misfunctions to precipitate neurodegeneration. We also recommend future research directions to validate such assertions for better understanding of disease mechanisms.
Collapse
Affiliation(s)
- Kevin Connolly
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University,Graduate Program in Molecular Biology, Cell Biology, and Biochemistry, Brown University
| | - Mikael Lehoux
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University
| | - Ryan O’Rourke
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University,Graduate Program in Pathobiology, Brown University
| | - Benedetta Assetta
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University
| | - Guzide Ayse Erdemir
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University
| | - Jack A Elias
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University,Department of Molecular Microbiology and Immunology, Brown University
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University
| | - Yu-Wen Alvin Huang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University,Department of Neurology, Warren Alpert Medical School of Brown University,Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University
| |
Collapse
|
23
|
Yuan H, Tang H, Shi L. Low expression of lncRNA UCA1 assists the diagnosis of type 2 diabetes mellitus and predicts an increased risk of cardiovascular complications. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2138561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Hui Yuan
- Department of Endocrinology, Daqing Oilfield General Hospital, Daqing, People’s Republic of China
| | - Haiyan Tang
- Department of Infectious Diseases, Daqing Oilfield General Hospital, Daqing, People’s Republic of China
| | - Lili Shi
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
24
|
Di Francesco AM, Verrecchia E, Manna S, Urbani A, Manna R. The chitinases as biomarkers in immune-mediate diseases. Clin Chem Lab Med 2022:cclm-2022-0767. [DOI: 10.1515/cclm-2022-0767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
Abstract
The role of chitinases has been focused as potential biomarkers in a wide number of inflammatory diseases, in monitoring active disease state, and predicting prognosis and response to therapies. The main chitinases, CHIT1 and YKL-40, are derived from 18 glycosyl hydrolases macrophage activation and play important roles in defense against chitin-containing pathogens and in food processing. Moreover, chitinases may have organ- as well as cell-specific effects in the context of infectious diseases and inflammatory disorders and able to induce tissue remodelling. The CHIT1 measurement is an easy, reproducible, reliable, and cost-effective affordable assay. The clinical use of CHIT1 for the screening of lysosomal storage disorders is quite practical, when proper cut-off values are determined for each laboratory. The potential of CHIT1 and chitinases has not been fully explored yet and future studies will produce many surprising discoveries in the immunology and allergology fields of research. However, since the presence of a null CHIT1 gene in a subpopulation would be responsible of false-negative values, the assay should be completed with the other markers such ACE and, if necessary, by genetic analysis when CHIT1 is unexpected low.
Collapse
Affiliation(s)
- Angela Maria Di Francesco
- Periodic Fever and Rare Diseases Research Centre, Catholic University of Sacred Heart , Rome , Italy
| | - Elena Verrecchia
- Periodic Fever and Rare Diseases Research Centre, Catholic University of Sacred Heart , Rome , Italy
| | - Stefano Manna
- Periodic Fever and Rare Diseases Research Centre, Catholic University of Sacred Heart , Rome , Italy
| | - Andrea Urbani
- Institute of Internal Medicine, Policlinico A. Gemelli Foundation IRCCS , Rome , Italy
- Department of Chemistry, Biochemistry and Molecular Biology , Policlinico A. Gemelli Foundation IRCCS , Rome , Italy
| | - Raffaele Manna
- Periodic Fever and Rare Diseases Research Centre, Catholic University of Sacred Heart , Rome , Italy
- Institute of Internal Medicine, Policlinico A. Gemelli Foundation IRCCS , Rome , Italy
| |
Collapse
|
25
|
Santos F, Cabreira V, Rocha S, Massano J. Blood Biomarkers for the Diagnosis of Neurodegenerative Dementia: A Systematic Review. J Geriatr Psychiatry Neurol 2022:8919887221141651. [PMID: 36423207 DOI: 10.1177/08919887221141651] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
IMPORTANCE Accurately diagnosing neurodegenerative dementia is often challenging due to overlapping clinical features. Disease specific biomarkers could enhance diagnostic accuracy. However, CSF analysis procedures and advanced imaging modalities are either invasive or high-priced, and routinely unavailable. Easily accessible disease biomarkers would be of utmost value for accurate differential diagnosis of dementia subtypes. OBJECTIVE To assess the diagnostic accuracy of blood-based biomarkers for the differential diagnosis of AD from Frontotemporal Lobar Degeneration (FTLD), or AD from Dementia with Lewy Bodies (DLB). METHODS Systematic review. Three databases (PubMed, Scopus, and Web of Science) were searched. Studies assessing blood-based biomarkers levels in AD versus FTLD, or AD versus DLB, and its diagnostic accuracy, were selected. When the same biomarker was assessed in three or more studies, a meta-analysis was performed. QUADAS-2 criteria were used for quality assessment. RESULTS Twenty studies were included in this analysis. Collectively, 905 AD patients were compared to 1262 FTLD patients, and 209 AD patients were compared to 246 DLB patients. Regarding biomarkers for AD versus FTLD, excellent discriminative accuracy (AUC >0.9) was found for p-tau181, p-tau217, synaptophysin, synaptopodin, GAP43 and calmodulin. Other biomarkers also demonstrated good accuracy (AUC = 0.8-0.9). For AD versus DLB distinction, only miR-21-5p and miR-451a achieved excellent accuracy (AUC >0.9). CONCLUSION Encouraging results were found for several biomarkers, alone or in combination. Prospective longitudinal designs and consensual protocols, comprising larger cohorts and homogeneous testing modalities across centres, are essential to validate the clinical value of blood biomarkers for the precise etiological diagnosis of dementia.
Collapse
Affiliation(s)
- Filipa Santos
- Department of Clinical Neurosciences and Mental Health, 26705Faculty of Medicine University of Porto, Porto, Portugal
| | - Verónica Cabreira
- Department of Clinical Neurosciences and Mental Health, 26705Faculty of Medicine University of Porto, Porto, Portugal.,Department of Neurology, 285211Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Sara Rocha
- iLoF - Intelligent Lab on Fiber, Oxford, UK.,Department of Biochemistry, 26705Faculty of Medicine University of Porto, Porto, Portugal
| | - João Massano
- Department of Clinical Neurosciences and Mental Health, 26705Faculty of Medicine University of Porto, Porto, Portugal.,Department of Neurology, 285211Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
26
|
Toward a Combination of Biomarkers for Molecular Characterization of Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms232214000. [PMID: 36430480 PMCID: PMC9695566 DOI: 10.3390/ijms232214000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system associated with chronic inflammation, demyelination, and axonal damage. MS is a highly heterogeneous disease that leads to discrepancies regarding the clinical appearance, progression, and therapy response of patients. Therefore, there is a strong unmet need for clinically relevant biomarkers capable of recapitulating the features of the disease. Experimental autoimmune encephalomyelitis (EAE) is a valuable model for studying the pathophysiology of MS as it recapitulates the main hallmarks of the disease: inflammation, blood-brain barrier (BBB) disruption, gliosis, myelin damage, and repair mechanisms. In this study, we used the EAE-PLP animal model and established a molecular RNA signature for each phase of the disease (onset, peak, remission). We compared variances of expression of known biomarkers by RT-qPCR in the brain and spinal cord of sham and EAE animals monitoring each of the five hallmarks of the disease. Using magnetic cell isolation technology, we isolated microglia and oligodendrocytes of mice of each category, and we compared the RNA expression variations. We identify genes deregulated during a restricted time frame, and we provide insight into the timing and interrelationships of pathological disease processes at the organ and cell levels.
Collapse
|
27
|
Zhou J, Benoit M, Sharoar MG. Recent advances in pre-clinical diagnosis of Alzheimer's disease. Metab Brain Dis 2022; 37:1703-1725. [PMID: 33900524 DOI: 10.1007/s11011-021-00733-4] [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: 01/02/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is the most common dementia with currently no known cures or disease modifying treatments (DMTs), despite much time and effort from the field. Diagnosis and intervention of AD during the early pre-symptomatic phase of the disease is thought to be a more effective strategy. Therefore, the detection of biomarkers has emerged as a critical tool for monitoring the effect of new AD therapies, as well as identifying patients most likely to respond to treatment. The establishment of the amyloid/tau/neurodegeneration (A/T/N) framework in 2018 has codified the contexts of use of AD biomarkers in neuroimaging and bodily fluids for research and diagnostic purposes. Furthermore, a renewed drive for novel AD biomarkers and innovative methods of detection has emerged with the goals of adding additional insight to disease progression and discovery of new therapeutic targets. The use of biomarkers has accelerated the development of AD drugs and will bring new therapies to patients in need. This review highlights recent methods utilized to diagnose antemortem AD.
Collapse
Affiliation(s)
- John Zhou
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
- Molecular Medicine Program, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Marc Benoit
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Md Golam Sharoar
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA.
| |
Collapse
|
28
|
Karayel O, Virreira Winter S, Padmanabhan S, Kuras YI, Vu DT, Tuncali I, Merchant K, Wills AM, Scherzer CR, Mann M. Proteome profiling of cerebrospinal fluid reveals biomarker candidates for Parkinson's disease. Cell Rep Med 2022; 3:100661. [PMID: 35732154 PMCID: PMC9245058 DOI: 10.1016/j.xcrm.2022.100661] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/29/2021] [Accepted: 05/23/2022] [Indexed: 11/16/2022]
Abstract
Parkinson's disease (PD) is a growing burden worldwide, and there is no reliable biomarker used in clinical routines to date. Cerebrospinal fluid (CSF) is routinely collected in patients with neurological symptoms and should closely reflect alterations in PD patients' brains. Here, we describe a scalable and sensitive mass spectrometry (MS)-based proteomics workflow for CSF proteome profiling. From two independent cohorts with over 200 individuals, our workflow reproducibly quantifies over 1,700 proteins from minimal CSF amounts. Machine learning determines OMD, CD44, VGF, PRL, and MAN2B1 to be altered in PD patients or to significantly correlate with clinical scores. We also uncover signatures of enhanced neuroinflammation in LRRK2 G2019S carriers, as indicated by increased levels of CTSS, PLD4, and HLA proteins. A comparison with our previously acquired urinary proteomes reveals a large overlap in PD-associated changes, including lysosomal proteins, opening up new avenues to improve our understanding of PD pathogenesis.
Collapse
Affiliation(s)
- Ozge Karayel
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Sebastian Virreira Winter
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
| | | | - Yuliya I Kuras
- APDA Center for Advanced Parkinson Research, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA; Precision Neurology Program, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Duc Tung Vu
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Idil Tuncali
- APDA Center for Advanced Parkinson Research, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA; Precision Neurology Program, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Kalpana Merchant
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anne-Marie Wills
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Clemens R Scherzer
- APDA Center for Advanced Parkinson Research, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA; Precision Neurology Program, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
29
|
Huang S, Wang YJ, Guo J. Biofluid Biomarkers of Alzheimer’s Disease: Progress, Problems, and Perspectives. Neurosci Bull 2022; 38:677-691. [PMID: 35306613 PMCID: PMC9206048 DOI: 10.1007/s12264-022-00836-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022] Open
Abstract
Since the establishment of the biomarker-based A-T-N (Amyloid/Tau/Neurodegeneration) framework in Alzheimer’s disease (AD), the diagnosis of AD has become more precise, and cerebrospinal fluid tests and positron emission tomography examinations based on this framework have become widely accepted. However, the A-T-N framework does not encompass the whole spectrum of AD pathologies, and problems with invasiveness and high cost limit the application of the above diagnostic methods aimed at the central nervous system. Therefore, we suggest the addition of an “X” to the A-T-N framework and a focus on peripheral biomarkers in the diagnosis of AD. In this review, we retrospectively describe the recent progress in biomarkers based on the A-T-N-X framework, analyze the problems, and present our perspectives on the diagnosis of AD.
Collapse
|
30
|
Morozova A, Zorkina Y, Abramova O, Pavlova O, Pavlov K, Soloveva K, Volkova M, Alekseeva P, Andryshchenko A, Kostyuk G, Gurina O, Chekhonin V. Neurobiological Highlights of Cognitive Impairment in Psychiatric Disorders. Int J Mol Sci 2022; 23:1217. [PMID: 35163141 PMCID: PMC8835608 DOI: 10.3390/ijms23031217] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
This review is focused on several psychiatric disorders in which cognitive impairment is a major component of the disease, influencing life quality. There are plenty of data proving that cognitive impairment accompanies and even underlies some psychiatric disorders. In addition, sources provide information on the biological background of cognitive problems associated with mental illness. This scientific review aims to summarize the current knowledge about neurobiological mechanisms of cognitive impairment in people with schizophrenia, depression, mild cognitive impairment and dementia (including Alzheimer's disease).The review provides data about the prevalence of cognitive impairment in people with mental illness and associated biological markers.
Collapse
Affiliation(s)
- Anna Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Yana Zorkina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Olga Abramova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Olga Pavlova
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Konstantin Pavlov
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Kristina Soloveva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Maria Volkova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Polina Alekseeva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Alisa Andryshchenko
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Georgiy Kostyuk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Olga Gurina
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
31
|
Pathophysiology of neurodegenerative diseases: An interplay among axonal transport failure, oxidative stress, and inflammation? Semin Immunol 2022; 59:101628. [PMID: 35779975 PMCID: PMC9807734 DOI: 10.1016/j.smim.2022.101628] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Neurodegenerative diseases (NDs) are heterogeneous neurological disorders characterized by a progressive loss of selected neuronal populations. A significant risk factor for most NDs is aging. Considering the constant increase in life expectancy, NDs represent a global public health burden. Axonal transport (AT) is a central cellular process underlying the generation and maintenance of neuronal architecture and connectivity. Deficits in AT appear to be a common thread for most, if not all, NDs. Neuroinflammation has been notoriously difficult to define in relation to NDs. Inflammation is a complex multifactorial process in the CNS, which varies depending on the disease stage. Several lines of evidence suggest that AT defect, axonopathy and neuroinflammation are tightly interlaced. However, whether these impairments play a causative role in NDs or are merely a downstream effect of neuronal degeneration remains unsettled. We still lack reliable information on the temporal relationship between these pathogenic mechanisms, although several findings suggest that they may occur early during ND pathophysiology. This article will review the latest evidence emerging on whether the interplay between AT perturbations and some aspects of CNS inflammation can participate in ND etiology, analyze their potential as therapeutic targets, and the urge to identify early surrogate biomarkers.
Collapse
|
32
|
Mavroudis I, Chowdhury R, Petridis F, Karantali E, Chatzikonstantinou S, Balmus IM, Luca IS, Ciobica A, Kazis D. YKL-40 as a Potential Biomarker for the Differential Diagnosis of Alzheimer's Disease. MEDICINA (KAUNAS, LITHUANIA) 2021; 58:medicina58010060. [PMID: 35056368 PMCID: PMC8777884 DOI: 10.3390/medicina58010060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder, associated with extensive neuronal loss, dendritic and synaptic changes resulting in significant cognitive impairment. An increased number of studies have given rise to the neuroinflammatory hypothesis in AD. It is widely accepted that AD brains show chronic inflammation, probably triggered by the presence of insoluble amyloid beta deposits and neurofibrillary tangles (NFT) and is also related to the activation of neuronal death cascade. In the present study we aimed to investigate the role of YKL-40 levels in the cerebrospinal fluid (CSF) in the diagnosis of AD, and to discuss whether there are further potential roles of this protein in the management and treatment of AD. We conducted an online search on PubMed, Web of Science, and the Cochrane library databases from 1990 to 2021. The quantitative analysis showed that the levels of YKL-40 were significantly higher in Alzheimer’s disease compared to controls, to mild cognitive impairment (MCI) AD (MCI-AD) and to stable MCI. They were also increased in MCI-AD compared to stable MCI. The present study shows that the CSF levels of YKL-40 could be potentially used as a biomarker for the prognosis of mild cognitive impairment and the likelihood of progression to AD, as well as for the differential diagnosis between AD and MCI.
Collapse
Affiliation(s)
- Ioannis Mavroudis
- Department of Neurology, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK; (I.M.); (R.C.)
| | - Rumana Chowdhury
- Department of Neurology, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK; (I.M.); (R.C.)
| | - Foivos Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Eleni Karantali
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Symela Chatzikonstantinou
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Ioana Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, ”Alexandru Ioan Cuza” University of Iasi, Alexandru Lapsuneanu Street, No. 26, 700057 Iasi, Romania;
| | - Iuliana Simona Luca
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No. 20A, 700505 Iasi, Romania
- Correspondence: (I.S.L.); (A.C.)
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No. 20A, 700505 Iasi, Romania
- Correspondence: (I.S.L.); (A.C.)
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| |
Collapse
|
33
|
Dönder A, Özdemir HH. Serum YKL-40 levels in patients with multiple sclerosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2021; 79:795-798. [PMID: 34669813 DOI: 10.1590/0004-282x-anp-2020-0326] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/15/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory disease affecting the central nervous system. The YKL-40 protein, which is secreted from various cells that contribute to inflammation and infection, plays a role in immune regulation. OBJECTIVE This study investigated the serum YKL-40 levels of patients with clinically isolated syndrome (CIS) and MS. METHODS The participants was divided into three groups: 1) patients with CIS (n = 20); 2) patients with relapsing-remitting MS (RRMS; n = 39); and 3) healthy individuals (n = 35). The YKL-40 levels in serum samples obtained from the participants were measured using enzyme-linked immunoassays. RESULTS The median serum YKL-40 level was 20.2 ng/mL (range 9.8-75.9 ng/mL) in the patients with CIS, 22.7 ng/mL (range 13.4-57.9 ng/mL) in the patients with RRMS and 11.0 ng/mL (range 10.0-17.3 ng/mL) in the control group (p < 0.001). The serum YKL-40 levels in the patients with RRMS were correlated with the patients' expanded disability status scale scores and ages (p < 0.05). No relationships were determined between the serum YKL-40 levels and the other variables (p > 0.05). The serum YKL-40 levels were higher in the CIS group than in the MS group. These findings show that the serum YKL-40 levels were high even at the beginning of the disease. The serum YKL-40 levels were also not involved in the progression to clinically definite MS. CONCLUSIONS The findings from this study suggested that YKL-40 may be a useful marker for the inflammatory process of MS.
Collapse
Affiliation(s)
- Ahmet Dönder
- Mardin Artuklu University, Vocational School of Health Services, Department of Medical Laboratory, Mardin, Turkey
| | | |
Collapse
|
34
|
Calsolaro V, Matthews PM, Donat CK, Livingston NR, Femminella GD, Guedes SS, Myers J, Fan Z, Tyacke RJ, Venkataraman AV, Perneczky R, Gunn R, Rabiner EA, Gentleman S, Parker CA, Murphy PS, Wren PB, Hinz R, Sastre M, Nutt DJ, Edison P. Astrocyte reactivity with late-onset cognitive impairment assessed in vivo using 11C-BU99008 PET and its relationship with amyloid load. Mol Psychiatry 2021; 26:5848-5855. [PMID: 34267329 PMCID: PMC8758500 DOI: 10.1038/s41380-021-01193-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 05/16/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
11C-BU99008 is a novel positron emission tomography (PET) tracer that enables selective imaging of astrocyte reactivity in vivo. To explore astrocyte reactivity associated with Alzheimer's disease, 11 older, cognitively impaired (CI) subjects and 9 age-matched healthy controls (HC) underwent 3T magnetic resonance imaging (MRI), 18F-florbetaben and 11C-BU99008 PET. The 8 amyloid (Aβ)-positive CI subjects had higher 11C-BU99008 uptake relative to HC across the whole brain, but particularly in frontal, temporal, medial temporal and occipital lobes. Biological parametric mapping demonstrated a positive voxel-wise neuroanatomical correlation between 11C-BU99008 and 18F-florbetaben. Autoradiography using 3H-BU99008 with post-mortem Alzheimer's brains confirmed through visual assessment that increased 3H-BU99008 binding localised with the astrocyte protein glial fibrillary acid protein and was not displaced by PiB or florbetaben. This proof-of-concept study provides direct evidence that 11C-BU99008 can measure in vivo astrocyte reactivity in people with late-life cognitive impairment and Alzheimer's disease. Our results confirm that increased astrocyte reactivity is found particularly in cortical regions with high Aβ load. Future studies now can explore how clinical expression of disease varies with astrocyte reactivity.
Collapse
Affiliation(s)
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Cornelius K Donat
- Department of Brain Sciences, Imperial College London, London, UK
- Centre for Blast Injury Studies, Imperial College London, London, UK
| | | | | | | | - Jim Myers
- Department of Brain Sciences, Imperial College London, London, UK
| | - Zhen Fan
- Department of Brain Sciences, Imperial College London, London, UK
| | - Robin J Tyacke
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- German Centre for Neurodegenerative Disorders (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Roger Gunn
- Department of Brain Sciences, Imperial College London, London, UK
- Invicro, London, UK
| | | | - Steve Gentleman
- Department of Brain Sciences, Imperial College London, London, UK
| | - Christine A Parker
- Department of Brain Sciences, Imperial College London, London, UK
- GlaxoSmithKline, Stevenage, UK
| | | | | | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, London, UK
| | - David J Nutt
- Department of Brain Sciences, Imperial College London, London, UK
| | - Paul Edison
- Department of Brain Sciences, Imperial College London, London, UK.
- Cardiff University, Cardiff, Wales, United Kingdom.
| |
Collapse
|
35
|
A 2A Adenosine Receptor as a Potential Biomarker and a Possible Therapeutic Target in Alzheimer's Disease. Cells 2021; 10:cells10092344. [PMID: 34571993 PMCID: PMC8469578 DOI: 10.3390/cells10092344] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative pathologies. Its incidence is in dramatic growth in Western societies and there is a need of both biomarkers to support the clinical diagnosis and drugs for the treatment of AD. The diagnostic criteria of AD are based on clinical data. However, it is necessary to develop biomarkers considering the neuropathology of AD. The A2A receptor, a G-protein coupled member of the P1 family of adenosine receptors, has different functions crucial for neurodegeneration. Its activation in the hippocampal region regulates synaptic plasticity and in particular glutamate release, NMDA receptor activation and calcium influx. Additionally, it exerts effects in neuroinflammation, regulating the secretion of pro-inflammatory cytokines. In AD patients, its expression is increased in the hippocampus/entorhinal cortex more than in the frontal cortex, a phenomenon not observed in age-matched control brains, indicating an association with AD pathology. It is upregulated in peripheral blood cells of patients affected by AD, thus reflecting its increase at central neuronal level. This review offers an overview on the main AD biomarkers and the potential role of A2A adenosine receptor as a new marker and therapeutic target.
Collapse
|
36
|
Prins S, Zhuparris A, Hart EP, Doll RJ, Groeneveld GJ. A cross-sectional study in healthy elderly subjects aimed at development of an algorithm to increase identification of Alzheimer pathology for the purpose of clinical trial participation. ALZHEIMERS RESEARCH & THERAPY 2021; 13:132. [PMID: 34274005 PMCID: PMC8286577 DOI: 10.1186/s13195-021-00874-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/04/2021] [Indexed: 11/10/2022]
Abstract
Background In the current study, we aimed to develop an algorithm based on biomarkers obtained through non- or minimally invasive procedures to identify healthy elderly subjects who have an increased risk of abnormal cerebrospinal fluid (CSF) amyloid beta42 (Aβ) levels consistent with the presence of Alzheimer’s disease (AD) pathology. The use of the algorithm may help to identify subjects with preclinical AD who are eligible for potential participation in trials with disease modifying compounds being developed for AD. Due to this pre-selection, fewer lumbar punctures will be needed, decreasing overall burden for study subjects and costs. Methods Healthy elderly subjects (n = 200; age 65–70 (N = 100) and age > 70 (N = 100)) with an MMSE > 24 were recruited. An automated central nervous system test battery was used for cognitive profiling. CSF Aβ1-42 concentrations, plasma Aβ1-40, Aβ1-42, neurofilament light, and total Tau concentrations were measured. Aβ1-42/1-40 ratio was calculated for plasma. The neuroinflammation biomarker YKL-40 and APOE ε4 status were determined in plasma. Different mathematical models were evaluated on their sensitivity, specificity, and positive predictive value. A logistic regression algorithm described the data best. Data were analyzed using a 5-fold cross validation logistic regression classifier. Results Two hundred healthy elderly subjects were enrolled in this study. Data of 154 subjects were used for the per protocol analysis. The average age of the 154 subjects was 72.1 (65–86) years. Twenty-four (27.3%) were Aβ positive for AD (age 65–83). The results of the logistic regression classifier showed that predictive features for Aβ positivity/negativity in CSF consist of sex, 7 CNS tests, and 1 plasma-based assay. The model achieved a sensitivity of 70.82% (± 4.35) and a specificity of 89.25% (± 4.35) with respect to identifying abnormal CSF in healthy elderly subjects. The receiver operating characteristic curve showed an AUC of 65% (± 0.10). Conclusion This algorithm would allow for a 70% reduction of lumbar punctures needed to identify subjects with abnormal CSF Aβ levels consistent with AD. The use of this algorithm can be expected to lower overall subject burden and costs of identifying subjects with preclinical AD and therefore of total study costs. Trial registration ISRCTN.org identifier: ISRCTN79036545 (retrospectively registered). Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00874-9.
Collapse
Affiliation(s)
- Samantha Prins
- Centre for Human drug Research, Leiden, the Netherlands.,Leiden University Medical Center, Leiden, the Netherlands
| | | | - Ellen P Hart
- Centre for Human drug Research, Leiden, the Netherlands
| | | | - Geert Jan Groeneveld
- Centre for Human drug Research, Leiden, the Netherlands. .,Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
37
|
Winder Z, Wilcock D, Jicha GA. Diagnostic and Prognostic Laboratory Testing for Alzheimer Disease. Clin Lab Med 2021; 40:289-303. [PMID: 32718500 DOI: 10.1016/j.cll.2020.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This article focuses on current clinical laboratory testing to diagnose Alzheimer disease and monitor its progression throughout its disease course. Several clinically available tests focus on analysis of amyloid and tau levels in cerebrospinal fluid as well as autosomal dominant and risk factor genes. Although the current armament of clinical laboratory testing is limited by invasiveness of cerebrospinal fluid collection, rarity of autosomal dominant genetic mutations, and uncertainties of risk inherent in nonpenetrant genes, the field is poised to advance the clinical repertoire of laboratory diagnostic testing.
Collapse
Affiliation(s)
- Zachary Winder
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA
| | - Donna Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA
| | - Gregory A Jicha
- Department of Neurology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA.
| |
Collapse
|
38
|
Cubas-Núñez L, Gil-Perotín S, Castillo-Villalba J, López V, Solís Tarazona L, Gasqué-Rubio R, Carratalá-Boscá S, Alcalá-Vicente C, Pérez-Miralles F, Lassmann H, Casanova B. Potential Role of CHI3L1+ Astrocytes in Progression in MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/3/e972. [PMID: 33658322 PMCID: PMC7931642 DOI: 10.1212/nxi.0000000000000972] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/21/2020] [Indexed: 11/15/2022]
Abstract
Objective Neurofilament light protein (NfL) and chitinase 3–like 1 (CHI3L1) are biomarkers for acute neuroaxonal damage and local inflammation, respectively. Thus, we set out to evaluate how these biomarkers were associated with clinical features of demyelinating diseases in parallel with the expression in brain autopsies from patients with similar disease stages, assuming their comparability. Methods NfL and CHI3L1 in CSF and serum CHI3L1 were assessed retrospectively in a cross-sectional cohort of controls (n = 17) and patients diagnosed with MS (n = 224), relapsing (n = 163) or progressive (n = 61); neuromyelitis optica (NMO, n = 7); and acute disseminated encephalomyelitis (ADEM, n = 15). Inflammatory activity was evaluated at the time of sampling, and CSF biomarker levels were related to the degree of inflammation in 22 brain autopsy tissues. Results During a clinical attack, the CSF NfL increased in MS, NMO, and ADEM, whereas CHI3L1 was only elevated in patients with NMO and ADEM and in outlier MS patients with extensive radiologic activity. Outside relapses, CHI3L1 levels only remained elevated in patients with progressive MS. CHI3L1 was detected in macrophages and astrocytes, predominantly in areas of active demyelination, and its expression by astrocytes in chronic lesions was independent of lymphocyte infiltrates and associated with active neurodegeneration. Conclusions Both CSF NfL and CHI3L1 augment during acute inflammation in demyelinating diseases. In MS, CHI3L1 may be associated with low-grade nonlymphocytic inflammation and active neurodegeneration and therefore linked to progressive disease. Classification of Evidence This study provides Class III evidence that CSF NfL and CHI3L1 levels increase in inflammatory brain diseases during acute inflammation.
Collapse
Affiliation(s)
- Laura Cubas-Núñez
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Sara Gil-Perotín
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria.
| | - Jéssica Castillo-Villalba
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Verónica López
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Luis Solís Tarazona
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Raquel Gasqué-Rubio
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Sara Carratalá-Boscá
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Carmen Alcalá-Vicente
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Francisco Pérez-Miralles
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Hans Lassmann
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| | - Bonaventura Casanova
- From the Neuroimmunology Unit (L.C.-N., S.G.-P., J.C.-V., V.L., R.G., S.C., C.A., F.P.-M., B.C.), Polytechnic and University Hospital La Fe; Neurology Department (L.S.T.), University Hospital Dr Peset, Valencia, Spain; and Department of Neuroimmunology (H.L.), Center for Brain Research, Vienna, Austria
| |
Collapse
|
39
|
Gonzales MM, Short MI, Satizabal CL, O’ Bryant S, Tracy RP, Zare H, Seshadri S. Blood biomarkers for dementia in Hispanic and non-Hispanic White adults. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12164. [PMID: 33860071 PMCID: PMC8033409 DOI: 10.1002/trc2.12164] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The study evaluated if blood markers reflecting diverse biological pathways differentiate clinical diagnostic groups among Hispanic and non-Hispanic White adults. METHODS Within Hispanic (n = 1193) and non-Hispanic White (n = 650) participants, serum total tau (t-tau), neurofilament light (NfL), ubiquitin carboxyl-terminal hydrolase LI, glial fibrillary acidic protein (GFAP), soluble cluster of differentiation-14, and chitinase-3-like protein 1 (YKL-40) were quantified. Mixed-effects partial proportional odds ordinal logistic regression and linear mixed-effects models were used to evaluate the association of biomarkers with diagnostic group and cognition, adjusting for age, sex, ethnicity, apolipoprotein E ε4, education, and site. RESULTS T-tau, NfL, GFAP, and YKL-40 discriminated between diagnostic groups (receiver operating curve: 0.647-0.873). Higher t-tau (odds ratio [OR] = 1.671, 95% confidence interval [CI] = 1.457-1.917, P < .001), NfL (OR = 2.150, 95% CI = 1.819-2.542, P < .001), GFAP (OR = 2.283, 95% CI = 1.915-2.722, P < .001), and YKL-40 (OR = 1.288, 95% CI = 1.125-1.475, P < .001) were associated with increased likelihood of dementia relative to cognitively unimpaired and mild cognitive impairment groups. Higher NfL was associated with poorer global cognition (β = -0.455, standard error [SE] = 0.083, P < .001), semantic fluency (β = -0.410, SE = 0.133, P = .002), attention/processing speed (β = 2.880, SE = 0.801, P < .001), and executive function (β = 5.965, SE = 2.037, P = .003). Higher GFAP was associated with poorer global cognition (β = -0.345, SE = 0.092, P = .001), learning (β = -1.426, SE = 0.359, P < .001), and memory (β = -0.890, SE = 0.266, P < .001). Higher YKL-40 (β = -0.537, SE = 0.186, P = .004) was associated with lower memory scores. Interactions with ethnicity were observed for learning (NfL, GFAP, YKL-40), memory (NfL, GFAP), and semantic fluency (NfL; interaction terms P < .008), which were generally no longer significant in a demographically matched subset of Hispanic and non-Hispanic White participants. DISCUSSION Blood biomarkers of neuronal/axonal and glial injury differentiated between clinical diagnostic groups in a bi-ethnic cohort of Hispanic and non-Hispanic Whites. Our results add to the growing literature indicating that blood biomarkers may be viable tools for detecting neurodegenerative conditions and highlight the importance of validation in diverse cohorts.
Collapse
Affiliation(s)
- Mitzi M. Gonzales
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of NeurologyUT Health San AntonioSan AntonioTexasUSA
| | - Meghan I. Short
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of Population Health SciencesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | - Sid O’ Bryant
- Institute for Translational Research and Department of Pharmacology & NeuroscienceUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Russel P. Tracy
- Departments of Pathology and Laboratory Medicine, and BiochemistryLarner College of Medicine, University of VermontBurlingtonVermontUSA
| | - Habil Zare
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of Cell Systems and AnatomyUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of NeurologyUT Health San AntonioSan AntonioTexasUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| |
Collapse
|
40
|
Shen H, Han C, Yang Y, Guo L, Sheng Y, Wang J, Li W, Zhai L, Wang G, Guan Q. Pyroptosis executive protein GSDMD as a biomarker for diagnosis and identification of Alzheimer's disease. Brain Behav 2021; 11:e02063. [PMID: 33587329 PMCID: PMC8035446 DOI: 10.1002/brb3.2063] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/01/2020] [Accepted: 01/17/2021] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE This study was mainly conducted to explore the expression changes of GSDMD and conventional markers (including T-Tau, Tau181p, and Aβ1-42 ) in the cerebrospinal fluid among patients with Alzheimer's disease (AD) and vascular dementia (VD), followed by determination of role of GSDMD in diagnosing and identifying AD and VD. METHODS In this study, 60 patients with VD, 60 patients with AD, and 50 healthy controls were enrolled. Lumbar puncture was performed to collect cerebrospinal fluid samples. Patients with VD and patients with AD were evaluated using the Mini-Mental State Examination (MMSE) scale, Montreal Cognitive Assessment (MoCA) scale, Clinical Dementia Rating (CDR) scale, Activity of Daily Living (ADL) scale, and Neuropsychiatric Inventory (NPI) questionnaire, aiming to determine the behavioral ability of patients. ELISA kit was purchased to determine the levels of GSDMD, T-Tau, Tau181p, and Aβ1-42 in cerebrospinal fluid, and the expression of inflammatory factors, IL-1β and IL-6, was also detected. RESULTS (1) The levels of GSDMD, T-Tau, and Tau181p in the cerebrospinal fluid were higher in patients with AD than those of patients with VD and healthy controls, while the levels of Aβ1-42 in the cerebrospinal fluid were lower in patients with AD than that in healthy controls and patients with VD. (2) GSDMD had good diagnostic accuracy in AD. Additionally, GSDMD, T-Tau, Tau181p, and Aβ1-42 had good discrimination accuracy in distinguishing AD and VD. (3) The expression levels of inflammatory factors (IL-1β and IL-6) in cerebrospinal fluid were higher in patients with AD than those of healthy controls and patients with VD, which were positively correlated with GSDMD expression. CONCLUSION The expression of GSDMD was increased in patients with AD, which could be used as a biomarker for AD diagnosis and identification from VD.
Collapse
Affiliation(s)
- Heping Shen
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chenyang Han
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yi Yang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Li Guo
- Department of Center Laboratory, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yongjia Sheng
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jin Wang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Wenyan Li
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Liping Zhai
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Genghuan Wang
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qiaobing Guan
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
41
|
Yang J, Jia L, Li Y, Qiu Q, Quan M, Jia J. Fluid Biomarkers in Clinical Trials for Alzheimer's Disease: Current and Future Application. J Alzheimers Dis 2021; 81:19-32. [PMID: 33749646 DOI: 10.3233/jad-201068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Alzheimer's disease (AD) research is entering a unique moment in which enormous information about the molecular basis of this disease is being translated into therapeutics. However, almost all drug candidates have failed in clinical trials over the past 30 years. These many trial failures have highlighted a need for the incorporation of biomarkers in clinical trials to help improve the trial design. Fluid biomarkers measured in cerebrospinal fluid and circulating blood, which can reflect the pathophysiological process in the brain, are becoming increasingly important in AD clinical trials. In this review, we first succinctly outline a panel of fluid biomarkers for neuropathological changes in AD. Then, we provide a comprehensive overview of current and future application of fluid biomarkers in clinical trials for AD. We also summarize the many challenges that have been encountered in efforts to integrate fluid biomarkers in clinical trials, and the barriers that have begun to be overcome. Ongoing research efforts in the field of fluid biomarkers will be critical to make significant progress in ultimately unveiling disease-modifying therapies in AD.
Collapse
Affiliation(s)
- Jianwei Yang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, People's Republic of China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Qiongqiong Qiu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, People's Republic of China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| |
Collapse
|
42
|
Ko PW, Lee HW, Lee M, Youn YC, Kim S, Kim JH, Kang K, Suk K. Increased plasma levels of chitinase 3-like 1 (CHI3L1) protein in patients with idiopathic normal-pressure hydrocephalus. J Neurol Sci 2021; 423:117353. [PMID: 33652290 DOI: 10.1016/j.jns.2021.117353] [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: 09/10/2020] [Revised: 01/22/2021] [Accepted: 02/15/2021] [Indexed: 10/22/2022]
Abstract
Idiopathic normal-pressure hydrocephalus (iNPH) is an uncommon neurological disorder with no known pathological hallmarks. INPH may share common degenerative pathways with other neurological diseases, such as Alzheimer's disease (AD). However, the reversible properties of iNPH may share differing pathophysiological mechanisms with other diseases. This study aimed at assessing the diagnostic value of plasma chitinase 3-like 1 (CHI3L1) protein levels as a disease-specific biomarker for iNPH. We selected both iNPH and AD patients as well as normal and disease control subjects from an enrolled dementia registry. A total of 121 AD, 80 iNPH, 13 idiopathic Parkinson's disease, and 23 mild cognitive impairment patients with 83 healthy controls were included in the final analysis. The Aβ42, total tau, and phosphorylated tau levels within the cerebrospinal fluid, as well as plasma levels of CHI3L1, were measured using commercially available enzyme-linked immunosorbent assay kits. CHI3L1 levels for iNPH patients were higher than those of the other groups. Analysis of covariance adjusting for age showed significantly increased plasma CHI3L1 levels in iNPH patients than in the controls (p < 0.001). CHI3L1 plasma levels may be useful in differentiating iNPH patients from healthy individuals.
Collapse
Affiliation(s)
- Pan-Woo Ko
- Department of Neurology, Daegu Health College Hospital, Daegu, Republic of Korea
| | - Ho-Won Lee
- Department of Neurology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Myunghoon Lee
- Research Center, D&P Biotech Inc, Seoul, Republic of Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Republic of Korea
| | - Jong-Heon Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Kyunghun Kang
- Department of Neurology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
43
|
McGrowder DA, Miller F, Vaz K, Nwokocha C, Wilson-Clarke C, Anderson-Cross M, Brown J, Anderson-Jackson L, Williams L, Latore L, Thompson R, Alexander-Lindo R. Cerebrospinal Fluid Biomarkers of Alzheimer's Disease: Current Evidence and Future Perspectives. Brain Sci 2021; 11:215. [PMID: 33578866 PMCID: PMC7916561 DOI: 10.3390/brainsci11020215] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease is a progressive, clinically heterogeneous, and particularly complex neurodegenerative disease characterized by a decline in cognition. Over the last two decades, there has been significant growth in the investigation of cerebrospinal fluid (CSF) biomarkers for Alzheimer's disease. This review presents current evidence from many clinical neurochemical studies, with findings that attest to the efficacy of existing core CSF biomarkers such as total tau, phosphorylated tau, and amyloid-β (Aβ42), which diagnose Alzheimer's disease in the early and dementia stages of the disorder. The heterogeneity of the pathophysiology of the late-onset disease warrants the growth of the Alzheimer's disease CSF biomarker toolbox; more biomarkers showing other aspects of the disease mechanism are needed. This review focuses on new biomarkers that track Alzheimer's disease pathology, such as those that assess neuronal injury (VILIP-1 and neurofilament light), neuroinflammation (sTREM2, YKL-40, osteopontin, GFAP, progranulin, and MCP-1), synaptic dysfunction (SNAP-25 and GAP-43), vascular dysregulation (hFABP), as well as CSF α-synuclein levels and TDP-43 pathology. Some of these biomarkers are promising candidates as they are specific and predict future rates of cognitive decline. Findings from the combinations of subclasses of new Alzheimer's disease biomarkers that improve their diagnostic efficacy in detecting associated pathological changes are also presented.
Collapse
Affiliation(s)
- Donovan A. McGrowder
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Fabian Miller
- Department of Physical Education, Faculty of Education, The Mico University College, 1A Marescaux Road, Kingston 5, Jamaica;
- Department of Biotechnology, Faculty of Science and Technology, The University of the West Indies, Kingston 7, Jamaica;
| | - Kurt Vaz
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Chukwuemeka Nwokocha
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| | - Cameil Wilson-Clarke
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| | - Melisa Anderson-Cross
- School of Allied Health and Wellness, College of Health Sciences, University of Technology, Kingston 7, Jamaica;
| | - Jabari Brown
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Lennox Anderson-Jackson
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Lowen Williams
- Department of Biotechnology, Faculty of Science and Technology, The University of the West Indies, Kingston 7, Jamaica;
| | - Lyndon Latore
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Rory Thompson
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Ruby Alexander-Lindo
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| |
Collapse
|
44
|
Ghantous CM, Kamareddine L, Farhat R, Zouein FA, Mondello S, Kobeissy F, Zeidan A. Advances in Cardiovascular Biomarker Discovery. Biomedicines 2020; 8:biomedicines8120552. [PMID: 33265898 PMCID: PMC7759775 DOI: 10.3390/biomedicines8120552] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are the leading causes of mortality worldwide. Among them, hypertension and its pathological complications pose a major risk for the development of other cardiovascular diseases, including heart failure and stroke. Identifying novel and early stage biomarkers of hypertension and other cardiovascular diseases is of paramount importance in predicting and preventing the major morbidity and mortality associated with these diseases. Biomarkers of such diseases or predisposition to their development are identified by changes in a specific indicator’s expression between healthy individuals and patients. These include changes in protein and microRNA (miRNA) levels. Protein profiling using mass spectrometry and miRNA screening utilizing microarray and sequencing have facilitated the discovery of proteins and miRNA as biomarker candidates. In this review, we summarized some of the different, promising early stage protein and miRNA biomarker candidates as well as the currently used biomarkers for hypertension and other cardiovascular diseases. Although a number of promising markers have been identified, it is unlikely that a single biomarker will unambiguously aid in the classification of these diseases. A multi-marker panel-strategy appears useful and promising for classifying and refining risk stratification among patients with cardiovascular disease.
Collapse
Affiliation(s)
- Crystal M. Ghantous
- Department of Nursing and Health Sciences, Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Keserwan 72, Lebanon;
| | - Layla Kamareddine
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Rima Farhat
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - Stefania Mondello
- Oasi Research Institute-IRCCS, 94018 Troina, Italy;
- Department of Biomedical and Dental Sciences and Morpho-functional Imaging, University of Messina, 98125 Messina, Italy
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - Asad Zeidan
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
- Department of Basic Medical Science, Faculty of Medicine, QU Health, Qatar University, Doha 2713, Qatar
- Correspondence: ; Tel.: +97-431-309-19
| |
Collapse
|
45
|
Ham HJ, Lee YS, Yun J, Son DJ, Lee HP, Han SB, Hong JT. K284-6111 alleviates memory impairment and neuroinflammation in Tg2576 mice by inhibition of Chitinase-3-like 1 regulating ERK-dependent PTX3 pathway. J Neuroinflammation 2020; 17:350. [PMID: 33222690 PMCID: PMC7681957 DOI: 10.1186/s12974-020-02022-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Background Alzheimer’s disease (AD) is one of the most prevalent neurodegenerative disorders characterized by gradual memory loss and neuropsychiatric symptoms. We have previously demonstrated that the 2-({3-[2-(1-cyclohexene-1-yl)ethyl]-6,7-dimethoxy-4-oxo-3,4-dihydro-2-quinazolinyl}sulfanyl)-N-(4-ethylphenyl)butanamide (K284-6111), the inhibitor of CHI3L1, has the inhibitory effect on memory impairment in Αβ infusion mouse model and on LPS-induced neuroinflammation in the murine BV-2 microglia and primary cultured astrocyte. Methods In the present study, we investigated the inhibitory effect of K284-6111 on memory dysfunction and neuroinflammation in Tg2576 transgenic mice, and a more detailed correlation of CHI3L1 and AD. To investigate the effects of K284-6111 on memory dysfunction, we administered K284-6111 (3 mg/kg, p.o.) daily for 4 weeks to Tg2576 mice, followed by behavioral tests of water maze test, probe test, and passive avoidance test. Results Administration of K284-6111 alleviated memory impairment in Tg2576 mice and had the effect of reducing the accumulation of Aβ and neuroinflammatory responses in the mouse brain. K284-6111 treatment also selectively inactivated ERK and NF-κB pathways, which were activated when CHI3L1 was overexpressed, in the mouse brain and in BV-2 cells. Web-based gene network analysis and our results of gene expression level in BV-2 cells showed that CHI3L1 is closely correlated with PTX3. Our result revealed that knockdown of PTX3 has an inhibitory effect on the production of inflammatory proteins and cytokines, and on the phosphorylation of ERK and IκBα. Conclusion These results suggest that K284-6111 could improve memory dysfunction by alleviating neuroinflammation through inhibiting CHI3L1 enhancing ERK-dependent PTX3 pathway.
Collapse
Affiliation(s)
- Hyeon Joo Ham
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Yong Sun Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
46
|
Gaetani L, Paolini Paoletti F, Bellomo G, Mancini A, Simoni S, Di Filippo M, Parnetti L. CSF and Blood Biomarkers in Neuroinflammatory and Neurodegenerative Diseases: Implications for Treatment. Trends Pharmacol Sci 2020; 41:1023-1037. [PMID: 33127098 DOI: 10.1016/j.tips.2020.09.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022]
Abstract
Neuroinflammatory and neurodegenerative diseases are characterized by the interplay of a number of molecular pathways that can be assessed through biofluids, especially cerebrospinal fluid and blood. Accordingly, the definition and classification of these disorders will move from clinical and pathological to biological criteria. The consequences of this biomarker-based diagnostic and prognostic approach are highly relevant to the field of drug development. Indeed, in view of the availability of disease-modifying drugs, fluid biomarkers offer a unique opportunity for improving the quality and applicability of results from clinical trials. Herein, we discuss the benefits of using fluid biomarkers for patient stratification, target engagement, and outcome assessment, as well as the most recent developments in neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Giovanni Bellomo
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Andrea Mancini
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Simone Simoni
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Lucilla Parnetti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy.
| |
Collapse
|
47
|
Advantages and Pitfalls in Fluid Biomarkers for Diagnosis of Alzheimer's Disease. J Pers Med 2020; 10:jpm10030063. [PMID: 32708853 PMCID: PMC7563364 DOI: 10.3390/jpm10030063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 01/08/2023] Open
Abstract
Alzheimer’s disease (AD) is a commonly occurring neurodegenerative disease in the advanced-age population, with a doubling of prevalence for each 5 years of age above 60 years. In the past two decades, there has been a sustained effort to find suitable biomarkers that may not only aide with the diagnosis of AD early in the disease process but also predict the onset of the disease in asymptomatic individuals. Current diagnostic evidence is supportive of some biomarker candidates isolated from cerebrospinal fluid (CSF), including amyloid beta peptide (Aβ), total tau (t-tau), and phosphorylated tau (p-tau) as being involved in the pathophysiology of AD. However, there are a few biomarkers that have been shown to be helpful, such as proteomic, inflammatory, oral, ocular and olfactory in the early detection of AD, especially in the individuals with mild cognitive impairment (MCI). To date, biomarkers are collected through invasive techniques, especially CSF from lumbar puncture; however, non-invasive (radio imaging) methods are used in practice to diagnose AD. In order to reduce invasive testing on the patients, present literature has highlighted the potential importance of biomarkers in blood to assist with diagnosing AD.
Collapse
|
48
|
Qin T, Prins S, Groeneveld GJ, Van Westen G, de Vries HE, Wong YC, Bischoff LJ, de Lange EC. Utility of Animal Models to Understand Human Alzheimer's Disease, Using the Mastermind Research Approach to Avoid Unnecessary Further Sacrifices of Animals. Int J Mol Sci 2020; 21:ijms21093158. [PMID: 32365768 PMCID: PMC7247586 DOI: 10.3390/ijms21093158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
To diagnose and treat early-stage (preclinical) Alzheimer’s disease (AD) patients, we need body-fluid-based biomarkers that reflect the processes that occur in this stage, but current knowledge on associated processes is lacking. As human studies on (possible) onset and early-stage AD would be extremely expensive and time-consuming, we investigate the potential value of animal AD models to help to fill this knowledge gap. We provide a comprehensive overview of processes associated with AD pathogenesis and biomarkers, current knowledge on AD-related biomarkers derived from on human and animal brains and body fluids, comparisons of biomarkers obtained in human AD and frequently used animal AD models, and emerging body-fluid-based biomarkers. In human studies, amyloid beta (Aβ), hyperphosphorylated tau (P-tau), total tau (T-tau), neurogranin, SNAP-25, glial fibrillary acidic protein (GFAP), YKL-40, and especially neurofilament light (NfL) are frequently measured. In animal studies, the emphasis has been mostly on Aβ. Although a direct comparison between human (familial and sporadic) AD and (mostly genetic) animal AD models cannot be made, still, in brain, cerebrospinal fluid (CSF), and blood, a majority of similar trends are observed for human AD stage and animal AD model life stage. This indicates the potential value of animal AD models in understanding of the onset and early stage of AD. Moreover, animal studies can be smartly designed to provide mechanistic information on the interrelationships between the different AD processes in a longitudinal fashion and may also include the combinations of different conditions that may reflect comorbidities in human AD, according to the Mastermind Research approach.
Collapse
Affiliation(s)
- Tian Qin
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
| | - Samantha Prins
- Centre for Human Drug Research (CHDR), 2333 CL Leiden, The Netherlands; (S.P.); (G.J.G.)
| | - Geert Jan Groeneveld
- Centre for Human Drug Research (CHDR), 2333 CL Leiden, The Netherlands; (S.P.); (G.J.G.)
| | - Gerard Van Westen
- Computational Drug Discovery, Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands;
| | - Helga E. de Vries
- Neuro-immunology research group, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands;
| | - Yin Cheong Wong
- Advanced Modelling and Simulation, UCB Celltech, Slough SL1 3WE, UK;
| | - Luc J.M. Bischoff
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
| | - Elizabeth C.M. de Lange
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
- Correspondence: ; Tel.: +31-71-527-6330
| |
Collapse
|
49
|
Zendehdel A, Roham M. Role of Helicobacter pylori infection in the manifestation of old age-related diseases. Mol Genet Genomic Med 2020; 8:e1157. [PMID: 32067423 PMCID: PMC7196471 DOI: 10.1002/mgg3.1157] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/10/2019] [Accepted: 01/11/2020] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori is one of the most prevalent infection worldwide. It affects individuals of different age groups. Elderly people tend to resist eradication treatment and worsening of infection can lead to several gastric and non-gastric pathologies. Aging-associated cellular and molecular alteration can increase the risk of other pathologies such as osteoporosis, Alzheimer's disease, Parkinson's disease, respiratory and renal dysfunction, and cancer in geriatric patients, more than other age groups. This review article highlights some of the most common old age diseases and the role of H. pylori infection as a risk factor to worsen the conditions, presented by the molecular evidences of these associations. These studies can help clinicians to understand the underlying pathogenesis of the disease and identify high-risk patients, aiding clearer diagnosis and treatment.
Collapse
Affiliation(s)
- Abolfazl Zendehdel
- Department of Geriatric MedicineZiaeian HospitalTehran University of Medical SciencesTehranIran
| | | |
Collapse
|
50
|
Moreno-Rodriguez M, Perez SE, Nadeem M, Malek-Ahmadi M, Mufson EJ. Frontal cortex chitinase and pentraxin neuroinflammatory alterations during the progression of Alzheimer's disease. J Neuroinflammation 2020; 17:58. [PMID: 32066474 PMCID: PMC7025403 DOI: 10.1186/s12974-020-1723-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/20/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chitinase 3-like 1 (CHI3L1), chitinase 3-like 2 (CHI3L2), and neuronal pentraxin II (NPTX2) are inflammatory biomarkers of Alzheimer's disease (AD). Although studies have demonstrated that cerebrospinal fluid levels of these proteins are changed in AD, no studies have undertaken a detailed examination of alterations in protein levels, cellular expression, and interaction with amyloid in the brain during the progression of AD. METHODS The study evaluated levels of both CHI3L1 and CHI3L2, NPTX2, ionized calcium-binding adapter molecule 1 (Iba1), complement component 1q (C1q), glial fibrillary acidic protein (GFAP), and CD44, in the frontal cortex of people who died with an antemortem clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), mild/moderate AD (mAD), and severe AD (sAD) using immunoblot and immunohistochemical techniques. RESULTS CHI3L1-immunoreactive (-ir) astrocyte numbers were increased in the frontal cortex and white matter in sAD compared to NCI. On the other hand, increases in GFAP and Iba1-ir cell numbers were observed in MCI compared to NCI but only in white matter. Western blot analyses revealed significantly lower frontal cortex CHI3L2 levels, whereas CD44 levels were increased in sAD. No significant differences for CHI3L1, GFAP, C1q, and NPTX2 protein levels were detected between clinical groups. Strong significant correlations were found between frontal cortex CHI3L1 and Iba1-ir cell numbers in white matter and CHI3L1 and C1q protein levels in the early stages of the disease. C1q and Iba1, CD44 with CHI3L2, and GFAP protein levels were associated during disease progression. CHI3L1 and Iba1 cell numbers in white matter showed a significant associations with episodic memory and perceptual speed. CONCLUSIONS White matter CHI3L1 inflammatory response is associated with cognitive impairment early in the onset of AD.
Collapse
Affiliation(s)
- Marta Moreno-Rodriguez
- Department of Neurobiology and Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, AZ, 85013, USA
| | - Sylvia E Perez
- Department of Neurobiology and Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, AZ, 85013, USA
| | - Muhammad Nadeem
- Department of Neurobiology and Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, AZ, 85013, USA
| | | | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, AZ, 85013, USA.
| |
Collapse
|