1
|
Han J, Hong R, Cao C, Zhang L, Sun A, Li Y, Chi Y, Zhang L, Yang Y, Qu X. Suppression of KDM5C mitigates the symptoms of Alzheimer's disease by up-regulating BDNF expression. Neurochem Int 2025; 186:105975. [PMID: 40180246 DOI: 10.1016/j.neuint.2025.105975] [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: 11/05/2024] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Histone methylation, a common form of chromatin remodeling, has been found to be associated with various neurological and cognitive disorders. However, little is known about how this mechanism contributes to the onset and progression of Alzheimer's disease (AD). Here, we found that lysine demethylase 5C (KDM5C), a histone H3 lysine 4 di- and tri-methyl (H3K4me2/3)-specific demethylase encoded by an X-linked mental retardation-related gene, displayed a progressive increase in the hippocampus with age in 3 × Tg-AD mice. Suppression of KDM5C partially mitigated the cognitive decline according to water maze, Y maze, and novel object recognition tests. In addition, significantly decreased amyloid plaques, enhanced long-term potentiation (LTP), and up-regulated expression of synaptic proteins were observed in KDM5C knockdown 3 × Tg-AD mice. Mechanistically, suppression of KDM5C could promote the expression of brain-derived neurotrophic factor (BDNF) to partially protect hippocampal neurons from beta-amyloid damage. In the promoter region of Bdnf, KDM5C was bound to the repressor element-1 (RE-1) motif to reduce the nearby H3K4me3 level and inhibit gene transcription. Mutations in the RE-1 motif reversed the inhibitory effect of KDM5C. Our results emphasize that KDM5C excess is one of the reasons for the onset and progression of AD and that suppression of KDM5C in the hippocampus should be considered a potential therapeutic target to ameliorate cognitive impairment and pathological symptoms in AD.
Collapse
Affiliation(s)
- Jingjing Han
- Department of Basic Medical Science, Jiangsu Medical College, Yancheng, 224005, Jiangsu, China
| | - Rui Hong
- Department of Basic Medical Science, Jiangsu Medical College, Yancheng, 224005, Jiangsu, China
| | - Cong Cao
- Department of Research and Education, The Fourth People's Hospital of Yancheng, Yancheng, 224000, Jiangsu, China
| | - Lina Zhang
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ao Sun
- Department of Pharmacology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, 224000, Jiangsu, China
| | - Yufei Li
- Department of Pharmacology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, 224000, Jiangsu, China
| | - Yinxiu Chi
- Department of Basic Medical Science, Jiangsu Medical College, Yancheng, 224005, Jiangsu, China
| | - Linlin Zhang
- Department of Pharmacology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, 224000, Jiangsu, China
| | - Ya Yang
- Group Health Section, The Affiliated Yancheng Maternity & Child Health Hospital of Yangzhou University, Yancheng, 224000, Jiangsu, China
| | - Xuebin Qu
- Department of Basic Medical Science, Jiangsu Medical College, Yancheng, 224005, Jiangsu, China.
| |
Collapse
|
2
|
Zoccali C, Capasso G. Genetic biomarkers of cognitive impairment and dementia of potential interest in CKD patients. J Nephrol 2024; 37:2473-2479. [PMID: 38970746 DOI: 10.1007/s40620-024-02006-6] [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: 04/16/2024] [Accepted: 06/15/2024] [Indexed: 07/08/2024]
Abstract
This review discusses genetic variants associated with cognitive dysfunction in chronic kidney disease (CKD) patients, emphasising the limited research in this area. Four studies have explored genetic markers of cognitive dysfunction in CKD, with findings suggesting shared genetic biomarkers between Alzheimer's Disease and CKD.Because of the limited specific research on genetic markers of cognitive dysfunction and dementia in CKD, we extracted data from the current literature studies on genetic markers in the general population that may be relevant to the CKD population. These markers include Apolipoprotein E (APOE), Complement Receptor 1 (CR1), Clusterin (CLU), Sortilin-related receptor 1 (SORL1), Catechol-O-methyltransferase (COMT), and Brain-derived neurotrophic factor (BDNF), all of which are known to be associated with cognitive dysfunction and dementia in other populations. These genes play various roles in lipid metabolism, inflammation, Aβ clearance, and neuronal function, making them potential candidates for studying cognitive decline in CKD patients.CKD-specific research is needed to understand the role of these genetic markers in CKD-related cognitive dysfunction. Investigating how these genes influence cognitive decline in CKD patients could provide valuable insights into early detection, targeted interventions, and personalised treatment strategies. Overall, genetic studies to enhance our understanding and management of cognitive dysfunction in CKD represent a clinical research priority in this population.
Collapse
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute, New York, USA.
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy.
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy.
| | - Giovambattista Capasso
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
3
|
Thomson D, Rosenich E, Maruff P, Lim YY. BDNF Val66Met moderates episodic memory decline and tau biomarker increases in early sporadic Alzheimer's disease. Arch Clin Neuropsychol 2024; 39:683-691. [PMID: 38454193 PMCID: PMC11345111 DOI: 10.1093/arclin/acae014] [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: 10/02/2023] [Revised: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
OBJECTIVE Allelic variation in the brain-derived neurotrophic factor (BDNF) Val66Met polymorphism has been shown to moderate rates of cognitive decline in preclinical sporadic Alzheimer's disease (AD; i.e., Aβ + older adults), and pre-symptomatic autosomal dominant Alzheimer's disease (ADAD). In ADAD, Met66 was also associated with greater increases in CSF levels of total-tau (t-tau) and phosphorylated tau (p-tau181). This study sought to determine the extent to which BDNF Val66Met is associated with changes in episodic memory and CSF t-tau and p-tau181 in Aβ + older adults in early-stage sporadic AD. METHOD Aβ + Met66 carriers (n = 94) and Val66 homozygotes (n = 192) enrolled in the Alzheimer's Disease Neuroimaging Initiative who did not meet criteria for AD dementia, and with at least one follow-up neuropsychological and CSF assessment, were included. A series of linear mixed models were conducted to investigate changes in each outcome over an average of 2.8 years, covarying for CSF Aβ42, APOE ε4 status, sex, age, baseline diagnosis, and years of education. RESULTS Aβ + Met66 carriers demonstrated significantly faster memory decline (d = 0.33) and significantly greater increases in CSF t-tau (d = 0.30) and p-tau181 (d = 0.29) compared to Val66 homozygotes, despite showing equivalent changes in CSF Aβ42. CONCLUSIONS These findings suggest that reduced neurotrophic support, which is associated with Met66 carriage, may increase vulnerability to Aβ-related tau hyperphosphorylation, neuronal dysfunction, and cognitive decline even prior to the emergence of dementia. Additionally, these findings highlight the need for neuropsychological and clinicopathological models of AD to account for neurotrophic factors and the genes which moderate their expression.
Collapse
Affiliation(s)
- Diny Thomson
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC 3168, Australia
| | | | - Paul Maruff
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC 3168, Australia
- Cogstate Ltd, Melbourne, VIC 3000, Australia
| | - Yen Ying Lim
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC 3168, Australia
| | | |
Collapse
|
4
|
Forsell P, Parrado Fernández C, Nilsson B, Sandin J, Nordvall G, Segerdahl M. Positive Allosteric Modulators of Trk Receptors for the Treatment of Alzheimer's Disease. Pharmaceuticals (Basel) 2024; 17:997. [PMID: 39204102 PMCID: PMC11357672 DOI: 10.3390/ph17080997] [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: 06/17/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Neurotrophins are important regulators of neuronal and non-neuronal functions. As such, the neurotrophins and their receptors, the tropomyosin receptor kinase (Trk) family of receptor tyrosine kinases, has attracted intense research interest and their role in multiple diseases including Alzheimer's disease has been described. Attempts to administer neurotrophins to patients have been reported, but the clinical trials have so far have been hampered by side effects or a lack of clear efficacy. Thus, much of the focus during recent years has been on identifying small molecules acting as agonists or positive allosteric modulators (PAMs) of Trk receptors. Two examples of successful discovery and development of PAMs are the TrkA-PAM E2511 and the pan-Trk PAM ACD856. E2511 has been reported to have disease-modifying effects in preclinical models, whereas ACD856 demonstrates both a symptomatic and a disease-modifying effect in preclinical models. Both molecules have reached the stage of clinical development and were reported to be safe and well tolerated in clinical phase 1 studies, albeit with different pharmacokinetic profiles. These two emerging small molecules are interesting examples of possible novel symptomatic and disease-modifying treatments that could complement the existing anti-amyloid monoclonal antibodies for the treatment of Alzheimer's disease. This review aims to present the concept of positive allosteric modulators of the Trk receptors as a novel future treatment option for Alzheimer's disease and other neurodegenerative and cognitive disorders, and the current preclinical and clinical data supporting this new concept. Preclinical data indicate dual mechanisms, not only as cognitive enhancers, but also a tentative neurorestorative function.
Collapse
Affiliation(s)
- Pontus Forsell
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| | - Cristina Parrado Fernández
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| | - Boel Nilsson
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
| | - Johan Sandin
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| | - Gunnar Nordvall
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| | - Märta Segerdahl
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| |
Collapse
|
5
|
Xia Z, Prescott EE, Urbanek A, Wareing HE, King MC, Olerinyova A, Dakin H, Leah T, Barnes KA, Matuszyk MM, Dimou E, Hidari E, Zhang YP, Lam JYL, Danial JSH, Strickland MR, Jiang H, Thornton P, Crowther DC, Ohtonen S, Gómez-Budia M, Bell SM, Ferraiuolo L, Mortiboys H, Higginbottom A, Wharton SB, Holtzman DM, Malm T, Ranasinghe RT, Klenerman D, De S. Co-aggregation with Apolipoprotein E modulates the function of Amyloid-β in Alzheimer's disease. Nat Commun 2024; 15:4695. [PMID: 38824138 PMCID: PMC11144216 DOI: 10.1038/s41467-024-49028-z] [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] [Accepted: 05/21/2024] [Indexed: 06/03/2024] Open
Abstract
Which isoforms of apolipoprotein E (apoE) we inherit determine our risk of developing late-onset Alzheimer's Disease (AD), but the mechanism underlying this link is poorly understood. In particular, the relevance of direct interactions between apoE and amyloid-β (Aβ) remains controversial. Here, single-molecule imaging shows that all isoforms of apoE associate with Aβ in the early stages of aggregation and then fall away as fibrillation happens. ApoE-Aβ co-aggregates account for ~50% of the mass of diffusible Aβ aggregates detected in the frontal cortices of homozygotes with the higher-risk APOE4 gene. We show how dynamic interactions between apoE and Aβ tune disease-related functions of Aβ aggregates throughout the course of aggregation. Our results connect inherited APOE genotype with the risk of developing AD by demonstrating how, in an isoform- and lipidation-specific way, apoE modulates the aggregation, clearance and toxicity of Aβ. Selectively removing non-lipidated apoE4-Aβ co-aggregates enhances clearance of toxic Aβ by glial cells, and reduces secretion of inflammatory markers and membrane damage, demonstrating a clear path to AD therapeutics.
Collapse
Affiliation(s)
- Zengjie Xia
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Emily E Prescott
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Agnieszka Urbanek
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Hollie E Wareing
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Marianne C King
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Anna Olerinyova
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Helen Dakin
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
- Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Tom Leah
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Katy A Barnes
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Martyna M Matuszyk
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Eleni Dimou
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Eric Hidari
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Yu P Zhang
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Jeff Y L Lam
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - John S H Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
- SUPA School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, KY16 9SS, UK
| | - Michael R Strickland
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hong Jiang
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Thornton
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Sohvi Ohtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mireia Gómez-Budia
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Simon M Bell
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Rohan T Ranasinghe
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK.
| | - Suman De
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK.
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
6
|
Bortolozzi A, Fico G, Berk M, Solmi M, Fornaro M, Quevedo J, Zarate CA, Kessing LV, Vieta E, Carvalho AF. New Advances in the Pharmacology and Toxicology of Lithium: A Neurobiologically Oriented Overview. Pharmacol Rev 2024; 76:323-357. [PMID: 38697859 PMCID: PMC11068842 DOI: 10.1124/pharmrev.120.000007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 05/05/2024] Open
Abstract
Over the last six decades, lithium has been considered the gold standard treatment for the long-term management of bipolar disorder due to its efficacy in preventing both manic and depressive episodes as well as suicidal behaviors. Nevertheless, despite numerous observed effects on various cellular pathways and biologic systems, the precise mechanism through which lithium stabilizes mood remains elusive. Furthermore, there is recent support for the therapeutic potential of lithium in other brain diseases. This review offers a comprehensive examination of contemporary understanding and predominant theories concerning the diverse mechanisms underlying lithium's effects. These findings are based on investigations utilizing cellular and animal models of neurodegenerative and psychiatric disorders. Recent studies have provided additional support for the significance of glycogen synthase kinase-3 (GSK3) inhibition as a crucial mechanism. Furthermore, research has shed more light on the interconnections between GSK3-mediated neuroprotective, antioxidant, and neuroplasticity processes. Moreover, recent advancements in animal and human models have provided valuable insights into how lithium-induced modifications at the homeostatic synaptic plasticity level may play a pivotal role in its clinical effectiveness. We focused on findings from translational studies suggesting that lithium may interface with microRNA expression. Finally, we are exploring the repurposing potential of lithium beyond bipolar disorder. These recent findings on the therapeutic mechanisms of lithium have provided important clues toward developing predictive models of response to lithium treatment and identifying new biologic targets. SIGNIFICANCE STATEMENT: Lithium is the drug of choice for the treatment of bipolar disorder, but its mechanism of action in stabilizing mood remains elusive. This review presents the latest evidence on lithium's various mechanisms of action. Recent evidence has strengthened glycogen synthase kinase-3 (GSK3) inhibition, changes at the level of homeostatic synaptic plasticity, and regulation of microRNA expression as key mechanisms, providing an intriguing perspective that may help bridge the mechanistic gap between molecular functions and its clinical efficacy as a mood stabilizer.
Collapse
Affiliation(s)
- Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Giovanna Fico
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michael Berk
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Marco Solmi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michele Fornaro
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Joao Quevedo
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Carlos A Zarate
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Lars V Kessing
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Eduard Vieta
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Andre F Carvalho
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| |
Collapse
|
7
|
Ji S, Kang J, Han C, Xu X, Chen M, Chen J, Chhetri JK, Pan J, Chan P. Potential role of APOE ɛ4 allele as a modifier for the association of BDNF Val66Met polymorphisms and cognitive impairment in community-dwelling older adults. Front Aging Neurosci 2024; 16:1330193. [PMID: 38374884 PMCID: PMC10876185 DOI: 10.3389/fnagi.2024.1330193] [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: 10/31/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
Objective To determine whether the brain-derived neurotrophic factor (BDNF) Val66Met polymorphism is associated with cognitive impairment (CI) in community-dwelling Chinese older adults, and to investigate whether this relationship is modified by the Apolipoprotein E (APOE) ɛ4 allele. Methods The study is a secondary analysis of 703 participants aged ≥60 years randomly enrolled from the Beijing Longitudinal Study of Aging II prospective cohort. The education-adjusted Mini-Mental State Examination and the Clinical Dementia Rating Scale were used to measure the cognitive performance of the subjects. The main effects and interactions (additive and multiplicative) of the BDNF Met and the APOE ε4 alleles on CI were estimated by logistic regression models. Results In total, 84 out of 703 older adults aged ≥60 years old had CI. No significant difference was observed in the risk of CI between participants with the BDNF Met allele and that of subjects without the BDNF Met allele (p = 0.213; p = 0.164). Individuals carrying both the BDNF Met and APOE ε4 alleles had an almost 1.5-fold increased odds of CI compared with carriers of the BDNF Met allele but without the APOE ε4 allele. The additive association indicated a positive interaction of both BDNF Met and APOE ε4 alleles with wide CIs (p = 0.021; p = 0.018). Conclusion The results suggest that the APOE ε4 allele may be a potential modifier for the association of the BDNF Val66Met polymorphism with CI in community-dwelling older adults.
Collapse
Affiliation(s)
- Shaozhen Ji
- Department of Neurology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Kang
- Department of Neurology, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Chao Han
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xitong Xu
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Meijie Chen
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Chen
- Department of Geriatrics, Shenzhen Hospital, Peking University, Shenzhen, China
| | - Jagadish K Chhetri
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jing Pan
- Department of Neurology, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Piu Chan
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
- Department of Neurobiology, Neurology and Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Gujral S, Burns M, Erickson KI, Rofey D, Peiffer JJ, Laws SM, Brown B. Dose-response effects of exercise on mental health in community-dwelling older adults: Exploration of genetic moderators. Int J Clin Health Psychol 2024; 24:100443. [PMID: 38304020 PMCID: PMC10831729 DOI: 10.1016/j.ijchp.2024.100443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
Background/Objective (1) Examine the role of exercise intensity on mental health symptoms in a community-based sample of older adults. (2) Explore the moderating role of genetic variation in brain-derived neurotrophic factor (BDNF) and apolipoprotein E (APOE) on the effects of exercise on mental health symptoms. Method This study is a secondary analysis of a three-arm randomized controlled trial, comparing the effects of 6 months of high-intensity aerobic training vs. moderate-intensity aerobic training vs. a no-contact control group on mental health symptoms assessed using the Depression, Anxiety, and Stress Scale (DASS). The BDNF Val66Met polymorphism and APOE ε4 carrier status were explored as genetic moderators of exercise effects on mental health symptoms. Results The exercise intervention did not influence mental health symptoms. The BDNF Val66Met polymorphism did not moderate intervention effects on mental health symptoms. APOE ε4 carrier status moderated the effect of intervention group on perceived stress over 6 months, such that APOE ε4 carriers, but not non-carriers, in the high-intensity aerobic training group showed a decline in perceived stress over 6 months. Conclusions APOE ε4 carrier status may modify the benefits of high-intensity exercise on perceived stress such that APOE ε4 carriers show a greater decline in stress as a result of exercise relative to non-APOE ε4 carriers.
Collapse
Affiliation(s)
- Swathi Gujral
- School of Medicine, Department of Psychiatry, University of Pittsburgh, 3501 Forbes Ave Suite 520-17, Pittsburgh, PA 15213, USA
| | - Marcia Burns
- Department of Psychology, Indianapolis University, 402 North Blackford Street, LD 124, Indianapolis, IN 46202, USA
| | - Kirk I Erickson
- AdventHealth Research Institute, Neuroscience, Orlando, FL 32804, USA
- Department of Psychology, University of Pittsburgh, USA
| | - Dana Rofey
- School of Medicine, Department of Psychiatry, University of Pittsburgh, 3501 Forbes Ave Suite 520-17, Pittsburgh, PA 15213, USA
| | - Jeremiah J Peiffer
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch Western, Australia
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Belinda Brown
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch Western, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
9
|
Parrado Fernandez C, Juric S, Backlund M, Dahlström M, Madjid N, Lidell V, Rasti A, Sandin J, Nordvall G, Forsell P. Neuroprotective and Disease-Modifying Effects of the Triazinetrione ACD856, a Positive Allosteric Modulator of Trk-Receptors for the Treatment of Cognitive Dysfunction in Alzheimer's Disease. Int J Mol Sci 2023; 24:11159. [PMID: 37446337 DOI: 10.3390/ijms241311159] [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: 05/30/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
The introduction of anti-amyloid monoclonal antibodies against Alzheimer's disease (AD) is of high importance. However, even though treated patients show very little amyloid pathology, there is only a modest effect on the rate of cognitive decline. Although this effect can possibly increase over time, there is still a need for alternative treatments that will improve cognitive function in patients with AD. Therefore, the purpose of this study was to characterize the triazinetrione ACD856, a novel pan-Trk positive allosteric modulator, in multiple models to address its neuroprotective and potential disease-modifying effects. The pharmacological effect of ACD856 was tested in recombinant cell lines, primary cortical neurons, or animals. We demonstrate that ACD856 enhanced NGF-induced neurite outgrowth, increased the levels of the pre-synaptic protein SNAP25 in PC12 cells, and increased the degree of phosphorylated TrkB in SH-SY5Y cells. In primary cortical neurons, ACD856 led to increased levels of phospho-ERK1/2, showed a neuroprotective effect against amyloid-beta or energy-deprivation-induced neurotoxicity, and increased the levels of brain-derived neurotrophic factor (BDNF). Consequently, administration of ACD856 resulted in a significant increase in BDNF in the brains of 21 months old mice. Furthermore, repeated administration of ACD856 resulted in a sustained anti-depressant effect, which lasted up to seven days, suggesting effects that go beyond merely symptomatic effects. In conclusion, the results confirm ACD856 as a cognitive enhancer, but more importantly, they provide substantial in vitro and in vivo evidence of neuroprotective and long-term effects that contribute to neurotrophic support and increased neuroplasticity. Presumably, the described effects of ACD856 may improve cognition, increase resilience, and promote neurorestorative processes, thereby leading to a healthier brain in patients with AD.
Collapse
Affiliation(s)
- Cristina Parrado Fernandez
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, 171 77 Solna, Sweden
| | - Sanja Juric
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
| | - Maria Backlund
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
| | | | - Nather Madjid
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
| | | | - Azita Rasti
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
| | - Johan Sandin
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, 171 77 Solna, Sweden
| | - Gunnar Nordvall
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, 171 77 Solna, Sweden
| | - Pontus Forsell
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, 171 77 Solna, Sweden
| |
Collapse
|
10
|
Insel PS, Kumar A, Hansson O, Mattsson-Carlgren N. Genetic Moderation of the Association of β-Amyloid With Cognition and MRI Brain Structure in Alzheimer Disease. Neurology 2023; 101:e20-e29. [PMID: 37085326 PMCID: PMC10351305 DOI: 10.1212/wnl.0000000000207305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/03/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES There is considerable heterogeneity in the association between increasing β-amyloid (Aβ) pathology and early cognitive dysfunction in preclinical Alzheimer disease (AD). At this stage, some individuals show no signs of cognitive dysfunction, while others show clear signs of decline. The factors explaining this heterogeneity are particularly important for understanding progression in AD but remain largely unknown. In this study, we examined an array of genetic variants that may influence the relationships among Aβ, brain structure, and cognitive performance in 2 large cohorts. METHODS In 2,953 cognitively unimpaired participants from the Anti-Amyloid Treatment in Asymptomatic Alzheimer disease (A4) study, interactions between genetic variants and 18F-Florbetapir PET standardized uptake value ratio (SUVR) to predict the Preclinical Alzheimer Cognitive Composite (PACC) were assessed. Genetic variants identified in the A4 study were evaluated in the Alzheimer Disease Neuroimaging Initiative (ADNI, N = 527) for their association with longitudinal cognition and brain atrophy in both cognitively unimpaired participants and those with mild cognitive impairment. RESULTS In the A4 study, 4 genetic variants significantly moderated the association between Aβ load and cognition. Minor alleles of 3 variants were associated with additional decreases in PACC scores with increasing Aβ SUVR (rs78021285, β = -2.29, SE = 0.40, p FDR = 0.02, nearest gene ARPP21; rs71567499, β = -2.16, SE = 0.38, p FDR = 0.02, nearest gene PPARD; and rs10974405, β = -1.68, SE = 0.29, p FDR = 0.02, nearest gene GLIS3). The minor allele of rs7825645 was associated with less decrease in PACC scores with increasing Aβ SUVR (β = 0.71, SE = 0.13, p FDR = 0.04, nearest gene FGF20). The genetic variant rs76366637, in linkage disequilibrium with rs78021285, was available in both the A4 and ADNI. In the A4, rs76366637 was strongly associated with reduced PACC scores with increasing Aβ SUVR (β = -1.01, SE = 0.21, t = -4.90, p < 0.001). In the ADNI, rs76366637 was associated with accelerated cognitive decline (χ2 = 15.3, p = 0.004) and atrophy over time (χ2 = 26.8, p < 0.001), with increasing Aβ SUVR. DISCUSSION Patterns of increased cognitive dysfunction and accelerated atrophy due to specific genetic variation may explain some of the heterogeneity in cognition in preclinical and prodromal AD. The genetic variant near ARPP21 associated with lower cognitive scores in the A4 and accelerated cognitive decline and brain atrophy in the ADNI may help to identify those at the highest risk of accelerated progression of AD.
Collapse
Affiliation(s)
- Philip S Insel
- From the Clinical Memory Research Unit (P.S.I., A.K., O.H., N.M.-C.), Faculty of Medicine, Lund University, Sweden; Department of Psychiatry and Behavioral Sciences (P.S.I.), University of California, San Francisco; Memory Clinic (O.H.), Department of Neurology (N.M.-C.), Skåne University Hospital, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden.
| | - Atul Kumar
- From the Clinical Memory Research Unit (P.S.I., A.K., O.H., N.M.-C.), Faculty of Medicine, Lund University, Sweden; Department of Psychiatry and Behavioral Sciences (P.S.I.), University of California, San Francisco; Memory Clinic (O.H.), Department of Neurology (N.M.-C.), Skåne University Hospital, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit (P.S.I., A.K., O.H., N.M.-C.), Faculty of Medicine, Lund University, Sweden; Department of Psychiatry and Behavioral Sciences (P.S.I.), University of California, San Francisco; Memory Clinic (O.H.), Department of Neurology (N.M.-C.), Skåne University Hospital, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit (P.S.I., A.K., O.H., N.M.-C.), Faculty of Medicine, Lund University, Sweden; Department of Psychiatry and Behavioral Sciences (P.S.I.), University of California, San Francisco; Memory Clinic (O.H.), Department of Neurology (N.M.-C.), Skåne University Hospital, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| |
Collapse
|
11
|
Choi J, Choi SY, Hong Y, Han YE, Oh SJ, Lee B, Choi CW, Kim MS. The central administration of vitisin a, extracted from Vitis vinifera, improves cognitive function and related signaling pathways in a scopolamine-induced dementia model. Biomed Pharmacother 2023; 163:114812. [PMID: 37148861 DOI: 10.1016/j.biopha.2023.114812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023] Open
Abstract
Neurodegenerative disorders, such as Alzheimer's disease (AD), are characterized by cognitive function loss and progressive memory impairment. Vitis vinifera, which is consumed in the form of fruits and wines in various countries, contains several dietary stilbenoids that have beneficial effects on neuronal disorders related to cognitive impairment. However, few studies have investigated the hypothalamic effects of vitisin A, a resveratrol tetramer derived from V. vinifera stembark, on cognitive functions and related signaling pathways. In this study, we conducted in vitro, ex vivo, and in vivo experiments with multiple biochemical and molecular analyses to investigate its pharmaceutical effects on cognitive functions. Treatment with vitisin A increased cell viability and cell survival under H2O2-exposed conditions in a neuronal SH-SY5 cell line. Ex vivo experiments showed that vitisin A treatment restored the scopolamine-induced disruption of long-term potentiation (LTP) in the hippocampal CA3-CA1 synapse, indicating the restoration of synaptic mechanisms of learning and memory. Consistently, central administration of vitisin A ameliorated scopolamine-induced disruptions of cognitive and memory functions in C57BL/6 mice, as evidenced by Y-maze and passive avoidance tests. Further studies showed that vitisin A upregulates BDNF-CREB signaling in the hippocampus. Together, our findings suggest that vitisin A exhibits neuroprotective effects, at least partially, by upregulating BDNF-CREB signaling and LTP.
Collapse
Affiliation(s)
- Jeongyoon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Sung-Yun Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Yuni Hong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Young-Eun Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Bonggi Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Chun Whan Choi
- Natural Product Research Team, Gyeonggi Biocenter, Gyeonggido Business and Science Accelerator, Suwon, Republic of Korea.
| | - Min Soo Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| |
Collapse
|
12
|
Nordvall G, Forsell P, Sandin J. Neurotrophin-targeted therapeutics: A gateway to cognition and more? Drug Discov Today 2022; 27:103318. [PMID: 35850433 DOI: 10.1016/j.drudis.2022.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 02/09/2023]
Abstract
Neurotrophins, such as brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), are small proteins expressed in the brain and peripheral tissues, which regulate several key aspects of neuronal function, including neurogenesis, synaptic plasticity and neuroprotection, but also programmed cell death. This broad range of effects is a result of a complex downstream signaling pathway, with differential spatial and temporal activation patterns further diversifying their physiological effects. Alterations in neurotrophin levels, or known polymorphisms in neurotrophin genes, have been linked to a variety of disorders, including depression and Alzheimer's disease (AD). Historically, their therapeutic potential in these disorders has been hampered by the lack of suitable tool molecules for clinical studies. However, recent advancements have led to the development of new therapeutic candidates, which are now in clinical testing.
Collapse
|
13
|
Elsworthy RJ, Dunleavy C, Whitham M, Aldred S. Exercise for the prevention of Alzheimer's disease: Multiple pathways to promote non-amyloidogenic AβPP processing. AGING AND HEALTH RESEARCH 2022. [DOI: 10.1016/j.ahr.2022.100093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
14
|
Riphagen JM, van Hooren RWE, Kenis G, Verhey FRJ, Jacobs HIL. Distinct Patterns Link the BDNF Val66Met Polymorphism to Alzheimer's Disease Pathology. J Alzheimers Dis 2022; 88:447-453. [PMID: 35662115 DOI: 10.3233/jad-215353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The brain-derived neurotropic growth factor (BDNF) gene has been linked to dementia, inflammation, and Apolipoprotein E (APOE) ɛ4 status. We used cerebrospinal fluid (CSF) amyloid-β (Aβ)42 and phosphorylated tau (p-tau) to investigate associations with BDNF polymorphisms and modifications by APOE ɛ4 or inflammation in a memory clinic population (n = 114; subjective cognitive decline, mild cognitive impairment, Alzheimer's disease). We found distinct pathways to Alzheimer's disease pathology: Val-Met displayed lower CSF-Aβ 42 in APOE ɛ4+ carriers, independent of p-tau, while Val-Val displayed greater p-tau at higher IL-6 and sub-threshold Aβ 42. This may contribute to resolving some inconsistencies in the BDNF literature and provide possible inroads to specific Aβ and tau interventions depending on BDNF polymorphism.
Collapse
Affiliation(s)
- Joost M Riphagen
- Alzheimer Center Limburg, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Department of Radiology, Charlestown, MA, USA.,Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.,Faculty of Health, Medicine and Life Sciences; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Roy W E van Hooren
- Alzheimer Center Limburg, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.,Faculty of Health, Medicine and Life Sciences; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Gunter Kenis
- Faculty of Health, Medicine and Life Sciences; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Frans R J Verhey
- Alzheimer Center Limburg, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Heidi I L Jacobs
- Alzheimer Center Limburg, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.,Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.,Faculty of Health, Medicine and Life Sciences; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
15
|
Neuner SM, Telpoukhovskaia M, Menon V, O'Connell KMS, Hohman TJ, Kaczorowski CC. Translational approaches to understanding resilience to Alzheimer's disease. Trends Neurosci 2022; 45:369-383. [PMID: 35307206 PMCID: PMC9035083 DOI: 10.1016/j.tins.2022.02.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/07/2022] [Accepted: 02/23/2022] [Indexed: 10/18/2022]
Abstract
Individuals who maintain cognitive function despite high levels of Alzheimer's disease (AD)-associated pathology are said to be 'resilient' to AD. Identifying mechanisms underlying resilience represents an exciting therapeutic opportunity. Human studies have identified a number of molecular and genetic factors associated with resilience, but the complexity of these cohorts prohibits a complete understanding of which factors are causal or simply correlated with resilience. Genetically and phenotypically diverse mouse models of AD provide new and translationally relevant opportunities to identify and prioritize new resilience mechanisms for further cross-species investigation. This review will discuss insights into resilience gained from both human and animal studies and highlight future approaches that may help translate these insights into therapeutics designed to prevent or delay AD-related dementia.
Collapse
Affiliation(s)
- Sarah M Neuner
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kristen M S O'Connell
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Tufts University, School of Medicine, Graduate School of Biomedical Sciences, Boston, MA 02111, USA; The University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME 04469, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Catherine C Kaczorowski
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Tufts University, School of Medicine, Graduate School of Biomedical Sciences, Boston, MA 02111, USA; The University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME 04469, USA.
| |
Collapse
|
16
|
Seidler K, Barrow M. Intermittent fasting and cognitive performance - Targeting BDNF as potential strategy to optimise brain health. Front Neuroendocrinol 2022; 65:100971. [PMID: 34929259 DOI: 10.1016/j.yfrne.2021.100971] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/11/2021] [Accepted: 12/12/2021] [Indexed: 12/26/2022]
Abstract
Aging is the major risk factor for neurodegenerative diseases, accelerated by excessive calorie consumption and sedentary lifestyles. Bioenergetic challenges such as intermittent fasting (IF) have shown to promote lifespan and healthspan via an adaptive stress response. Activity-dependent brain-derived neurotrophic factor (BDNF) has emerged as key regulator of cognitive performance and brain health. This review aims to investigate the pathophysiological mechanisms linking IF and cognitive function with a focus on the role of BDNF, evaluating evidence from pre-clinical and human studies. A systematic literature search was performed. 82 peer-reviewed papers were accepted, critically appraised and summarised in a narrative analysis. Aging-related loss of BDNF has been associated with reduced synaptic plasticity, memory and learning as well as increased risk of cognitive impairment and Alzheimer's disease. IF was consistently reported to upregulate BDNF and improve cognitive performance in animal models. Further research is required to assess cognitive outcomes of IF in humans.
Collapse
Affiliation(s)
- Karin Seidler
- Centre for Nutrition Education and Lifestyle Management, Chapel Gardens, 14 Rectory Road, Wokingham RG40 1DH, United Kingdom.
| | - Michelle Barrow
- Centre for Nutrition Education and Lifestyle Management, Chapel Gardens, 14 Rectory Road, Wokingham RG40 1DH, United Kingdom
| |
Collapse
|
17
|
Golde TE. Alzheimer’s disease – the journey of a healthy brain into organ failure. Mol Neurodegener 2022; 17:18. [PMID: 35248124 PMCID: PMC8898417 DOI: 10.1186/s13024-022-00523-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/17/2022] [Indexed: 12/19/2022] Open
Abstract
As the most common dementia, Alzheimer’s disease (AD) exacts an immense personal, societal, and economic toll. AD was first described at the neuropathological level in the early 1900s. Today, we have mechanistic insight into select aspects of AD pathogenesis and have the ability to clinically detect and diagnose AD and underlying AD pathologies in living patients. These insights demonstrate that AD is a complex, insidious, degenerative proteinopathy triggered by Aβ aggregate formation. Over time Aβ pathology drives neurofibrillary tangle (NFT) pathology, dysfunction of virtually all cell types in the brain, and ultimately, overt neurodegeneration. Yet, large gaps in our knowledge of AD pathophysiology and huge unmet medical need remain. Though we largely conceptualize AD as a disease of aging, heritable and non-heritable factors impact brain physiology, either continuously or at specific time points during the lifespan, and thereby alter risk for devolvement of AD. Herein, I describe the lifelong journey of a healthy brain from birth to death with AD, while acknowledging the many knowledge gaps that remain regarding our understanding of AD pathogenesis. To ensure the current lexicon surrounding AD changes from inevitable, incurable, and poorly manageable to a lexicon of preventable, curable, and manageable we must address these knowledge gaps, develop therapies that have a bigger impact on clinical symptoms or progression of disease and use these interventions at the appropriate stage of disease.
Collapse
|
18
|
Kim J, Jung SH, Choe YS, Kim S, Kim B, Kim HR, Son SJ, Hong CH, Na DL, Kim HJ, Cho SJ, Won HH, Seo SW. Ethnic differences in the frequency of β-amyloid deposition in cognitively normal individuals. Neurobiol Aging 2022; 114:27-37. [DOI: 10.1016/j.neurobiolaging.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 10/18/2022]
|
19
|
Gao L, Zhang Y, Sterling K, Song W. Brain-derived neurotrophic factor in Alzheimer's disease and its pharmaceutical potential. Transl Neurodegener 2022; 11:4. [PMID: 35090576 PMCID: PMC8796548 DOI: 10.1186/s40035-022-00279-0] [Citation(s) in RCA: 218] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/01/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic abnormalities are a cardinal feature of Alzheimer's disease (AD) that are known to arise as the disease progresses. A growing body of evidence suggests that pathological alterations to neuronal circuits and synapses may provide a mechanistic link between amyloid β (Aβ) and tau pathology and thus may serve as an obligatory relay of the cognitive impairment in AD. Brain-derived neurotrophic factors (BDNFs) play an important role in maintaining synaptic plasticity in learning and memory. Considering AD as a synaptic disorder, BDNF has attracted increasing attention as a potential diagnostic biomarker and a therapeutical molecule for AD. Although depletion of BDNF has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation and neuronal apoptosis, the exact mechanisms underlying the effect of impaired BDNF signaling on AD are still unknown. Here, we present an overview of how BDNF genomic structure is connected to factors that regulate BDNF signaling. We then discuss the role of BDNF in AD and the potential of BDNF-targeting therapeutics for AD.
Collapse
Affiliation(s)
- Lina Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
20
|
Abanmy N, Alsabhan J, Gard P, Scutt G. Association between the Val66Met polymorphism (rs6265/G196A) of the BDNF gene and cognitive performance with SSRI use in Arab Alzheimer's disease patients. Saudi Pharm J 2022; 29:1392-1398. [PMID: 35002376 PMCID: PMC8720700 DOI: 10.1016/j.jsps.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
Brain derived neutrophic factor (BDNF) is a protein and a member of the neurotrophin family of growth factors, supports the survival of existing neurons and encourages the growth and differentiation of new neurons and synapses. The BDNF gene Val66Met polymorphism (rs6265/G196A) is responsible for BDNF synthesis that impact BDNF function that includes memory and cognition. This study investigated whether the BDNF gene Val66Met polymorphism (rs6265/G196A) is associated with cognitive function changes in both Alzheimer disease (AD) patients and elderly participants. In addition the impact of SSRI use on cognition improvement will be assessed. Healthy young, middle ages (25–59 years old) and elderly (more than 60 years old) participants (140) as well as 40 AD patients of whom are both of Saudi Arabian origin were recruited. The genotyping for the association study was performed by real-time PCR using Taqman chemistry in the ABI Prism 7900HT Sequence Detection System. Both Mini-Mental Status Examination (MMSE) and Clinical Dementia Rating (CDR) were used to assess cognitive function of healthy and AD participants, respectively. The findings showed that the BDNF Val66Met genotype distributions and allele frequencies have significant association with cognitive performance in both elderly control group and AD patients. The main findings showed that carriers of GG homozygotes (Val/Val) have superior cognitive performance among AD patients and elderly control subjects. In addition the use of SSRIs in 13 AD patients and 17 elderly participants positively improved cognitive function in elderly (p > 0.001) but not in AD patients (p = 0.1).
Collapse
Affiliation(s)
- N Abanmy
- College of Pharmacy, King Saud University, Riyadh 14212, Saudi Arabia
| | - J Alsabhan
- College of Pharmacy, King Saud University, Riyadh 14212, Saudi Arabia
| | - P Gard
- School of Pharmacy and biomolecular sciences, University of Brighton, Brighton BN2 4AT, UK
| | - G Scutt
- School of Pharmacy and biomolecular sciences, University of Brighton, Brighton BN2 4AT, UK
| |
Collapse
|
21
|
Therapeutic potential of neurotrophic factors in Alzheimer's Disease. Mol Biol Rep 2021; 49:2345-2357. [PMID: 34826049 DOI: 10.1007/s11033-021-06968-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia among the elderly population. AD is accompanied with the dysregulation of specific neurotrophic factors (NTFs) and their receptors, which plays a critical role in neuronal degeneration. NTFs are small proteins with therapeutic potential for human neurodegenerative diseases. These growth factors are categorized into four families: neurotrophins, neurokines, the glial cell line-derived NTF family of ligands, and the newly discovered cerebral dopamine NTF/mesencephalic astrocyte-derived NTF family. NTFs are capable of preventing cell death in degenerative conditions and can increase the neuronal growth and function in these disorders. Nevertheless, the adverse side effects of NTFs delivery and poor diffusion of these factors in the brain restrict the efficacy of NTFs therapy in clinical situations. MATERIALS AND METHODS In this review, we focus on the current advances in the use of NTFs to treat AD and summarize previous experimental and clinical studies for supporting the protective and therapeutic effects of these factors. CONCLUSION Based on reports, NTFs are considered as new and promising candidates for treating AD and AD-associated cognitive impairment.
Collapse
|
22
|
Souto JJ, Silva GM, Almeida NL, Shoshina II, Santos NA, Fernandes TP. Age-related episodic memory decline and the role of amyloid-β: a systematic review. Dement Neuropsychol 2021; 15:299-313. [PMID: 34630918 PMCID: PMC8485646 DOI: 10.1590/1980-57642021dn15-030002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/22/2021] [Indexed: 12/25/2022] Open
Abstract
Aging has been associated with the functional decline of episodic memory (EM). Unanswered questions are whether the decline of EM occurs even during healthy aging and whether this decline is related to amyloid-β (Aβ) deposition in the hippocampus. Objective The main purpose of this study was to investigate data on the relationship between the age-related EM decline and Aβ deposition. Methods We searched the Cochrane, MEDLINE, Scopus, and Web of Science databases and reference lists of retrieved articles that were published in the past 10 years. The initial literature search identified 517 studies. After screening the title, abstract, key words, and reference lists, 56 studies met the inclusion criteria. Results The overall results revealed that increases in Aβ are related to lower hippocampal volume and worse performance on EM tests. The results of this systematic review revealed that high levels of Aβ may be related to EM deficits and the progression to Alzheimer's disease. Conclusions We discussed the strengths and pitfalls of various tests and techniques used for investigating EM and Aβ deposition, methodological issues, and potential directions for future research.
Collapse
Affiliation(s)
- Jandirlly Julianna Souto
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| | - Gabriella Medeiros Silva
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| | - Natalia Leandro Almeida
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| | | | - Natanael Antonio Santos
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| | - Thiago Paiva Fernandes
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| |
Collapse
|
23
|
van den Bosch KA, Verberk IMW, Ebenau JL, van der Lee SJ, Jansen IE, Prins ND, Scheltens P, Teunissen CE, Van der Flier WM. BDNF-Met polymorphism and amyloid-beta in relation to cognitive decline in cognitively normal elderly: the SCIENCe project. Neurobiol Aging 2021; 108:146-154. [PMID: 34601245 DOI: 10.1016/j.neurobiolaging.2021.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 11/25/2022]
Abstract
Brain-derived neurotrophic factor (BNDF) plays a role in synapse integrity. We investigated in 398 cognitively normal adults (60±8years, 41% female, MMSE=28±1) the joint association of the Val66Met polymorphism of the BDNF gene (Met+/-) and plasma BDNF levels and abnormal cerebrospinal fluid (CSF) amyloid-beta status (A+/-) with cognitive decline and dementia risk. Age-, sex- and education-adjusted linear mixed models showed that compared to Met-A-, Met+A+ showed steeper decline on tests of global cognition, memory, language, attention and executive functioning, while Met-A+ showed steeper decline on a smaller number of tests. There were no associations between Met+A- and cognitive decline. Cox models showed that compared to Met-A-, Met+A+ participants were at increased risk of dementia (HR=8.8, 95%CI: 2.8-27.9), as were Met-A+ participants (HR=6.5, 95%CI: 2.2-19.5). Lower plasma BDNF was associated with an increased risk of progression to dementia in the A+ participants. Our results imply that Met-carriage on top of amyloid-beta pathology might increase rate of cognitive decline to dementia.
Collapse
Affiliation(s)
- Karlijn A van den Bosch
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Inge M W Verberk
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Neurochemistry Laboratory, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Jarith L Ebenau
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Iris E Jansen
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, VU, Amsterdam, The Netherlands
| | - Niels D Prins
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Brain Research Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Neurochemistry Laboratory, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M Van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Department of Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Therriault J, Benedet AL, Pascoal TA, Mathotaarachchi S, Savard M, Chamoun M, Thomas E, Kang MS, Lussier F, Tissot C, Soucy JP, Massarweh G, Rej S, Saha-Chaudhuri P, Poirier J, Gauthier S, Rosa-Neto P. APOEε4 potentiates the relationship between amyloid-β and tau pathologies. Mol Psychiatry 2021; 26:5977-5988. [PMID: 32161362 PMCID: PMC8758492 DOI: 10.1038/s41380-020-0688-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 01/06/2020] [Accepted: 02/12/2020] [Indexed: 11/08/2022]
Abstract
APOEε4 is the most well-established genetic risk factor for sporadic Alzheimer's disease and is associated with cerebral amyloid-β. However, the association between APOEε4 and tau pathology, the other major proteinopathy of Alzheimer's disease, has been controversial. Here, we sought to determine whether the relationship between APOEε4 and tau pathology is determined by local interactions with amyloid-β. We examined three independent samples of cognitively unimpaired, mild cognitive impairment and Alzheimer's disease subjects: (1) 211 participants who underwent tau-PET with [18F]MK6240 and amyloid-PET with [18F]AZD4694, (2) 264 individuals who underwent tau-PET with [18F]Flortaucipir and amyloid-PET with [18F]Florbetapir and (3) 487 individuals who underwent lumbar puncture and amyloid-PET with [18F]Florbetapir. Using a novel analytical framework, we applied voxel-wise regression models to assess the interactive effect of APOEε4 and amyloid-β on tau load, independently of age and clinical diagnosis. We found that the interaction effect between APOEε4 and amyloid-β, rather than the sum of their independent effects, was related to increased tau load in Alzheimer's disease-vulnerable regions. The interaction between one APOEε4 allele and amyloid-β was related to increased tau load, while the interaction between amyloid-β and two APOEε4 alleles was related to a more widespread pattern of tau aggregation. Our results contribute to an emerging framework in which the elevated risk of developing dementia conferred by APOEε4 genotype involves mechanisms associated with both amyloid-β and tau aggregation. These results may have implications for future disease-modifying therapeutic trials targeting amyloid or tau pathologies.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Andrea L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Emilie Thomas
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Firoza Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Cecile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Gassan Massarweh
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Radiochemistry, McGill University, Montreal, QC, Canada
| | - Soham Rej
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | | | - Judes Poirier
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montreal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Montreal Neurological Institute, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
25
|
The potential roles of genetic factors in predicting ageing-related cognitive change and Alzheimer's disease. Ageing Res Rev 2021; 70:101402. [PMID: 34242808 DOI: 10.1016/j.arr.2021.101402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/22/2021] [Accepted: 07/02/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder of uncertain aetiology, although substantial research has been conducted to explore important factors related to risk of onset and progression. Both lifestyle (e.g., complex mental stimulation, vascular health) and genetic factors (e.g., APOE, BDNF, PICALM, CLU, APP, PSEN1, PSEN2, and other genes) have been associated with AD risk. Despite more than thirty years of genetic research, much of the heritability of AD is not explained by measured loci. This suggests that the missing heritability of AD might be potentially related to rare variants, gene-environment and gene-gene interactions, and potentially epigenetic modulators. Moreover, while ageing is the most substantial factor risk for AD, there are limited longitudinal studies examining the association of genetic factors with decline in cognitive function due to ageing and the preclinical stages of this condition. This review summarises findings from currently available research on the genetic factors of ageing-related cognitive change and AD and suggests some future research directions.
Collapse
|
26
|
Puglisi-Allegra S, Ruggieri S, Fornai F. Translational evidence for lithium-induced brain plasticity and neuroprotection in the treatment of neuropsychiatric disorders. Transl Psychiatry 2021; 11:366. [PMID: 34226487 PMCID: PMC8257731 DOI: 10.1038/s41398-021-01492-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence indicates lithium (Li+) efficacy in neuropsychiatry, pointing to overlapping mechanisms that occur within distinct neuronal populations. In fact, the same pathway depending on which circuitry operates may fall in the psychiatric and/or neurological domains. Li+ restores both neurotransmission and brain structure unveiling that psychiatric and neurological disorders share common dysfunctional molecular and morphological mechanisms, which may involve distinct brain circuitries. Here an overview is provided concerning the therapeutic/neuroprotective effects of Li+ in different neuropsychiatric disorders to highlight common molecular mechanisms through which Li+ produces its mood-stabilizing effects and to what extent these overlap with plasticity in distinct brain circuitries. Li+ mood-stabilizing effects are evident in typical bipolar disorder (BD) characterized by a cyclic course of mania or hypomania followed by depressive episodes, while its efficacy is weaker in the opposite pattern. We focus here on neural adaptations that may underlie psychostimulant-induced psychotic development and to dissect, through the sensitization process, which features are shared in BD and other psychiatric disorders, including schizophrenia. The multiple functions of Li+ highlighted here prove its exceptional pharmacology, which may help to elucidate its mechanisms of action. These may serve as a guide toward a multi-drug strategy. We propose that the onset of sensitization in a specific BD subtype may predict the therapeutic efficacy of Li+. This model may help to infer in BD which molecular mechanisms are relevant to the therapeutic efficacy of Li+.
Collapse
Affiliation(s)
| | | | - Francesco Fornai
- IRCCS Neuromed, Via Atinense 18, 86077, Pozzilli (IS), Italy.
- Human Anatomy, Department of Translational Research and New technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa (PI), Italy.
| |
Collapse
|
27
|
Fowler C, Rainey-Smith SR, Bird S, Bomke J, Bourgeat P, Brown BM, Burnham SC, Bush AI, Chadunow C, Collins S, Doecke J, Doré V, Ellis KA, Evered L, Fazlollahi A, Fripp J, Gardener SL, Gibson S, Grenfell R, Harrison E, Head R, Jin L, Kamer A, Lamb F, Lautenschlager NT, Laws SM, Li QX, Lim L, Lim YY, Louey A, Macaulay SL, Mackintosh L, Martins RN, Maruff P, Masters CL, McBride S, Milicic L, Peretti M, Pertile K, Porter T, Radler M, Rembach A, Robertson J, Rodrigues M, Rowe CC, Rumble R, Salvado O, Savage G, Silbert B, Soh M, Sohrabi HR, Taddei K, Taddei T, Thai C, Trounson B, Tyrrell R, Vacher M, Varghese S, Villemagne VL, Weinborn M, Woodward M, Xia Y, Ames D. Fifteen Years of the Australian Imaging, Biomarkers and Lifestyle (AIBL) Study: Progress and Observations from 2,359 Older Adults Spanning the Spectrum from Cognitive Normality to Alzheimer's Disease. J Alzheimers Dis Rep 2021; 5:443-468. [PMID: 34368630 PMCID: PMC8293663 DOI: 10.3233/adr-210005] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: The Australian Imaging, Biomarkers and Lifestyle (AIBL) Study commenced in 2006 as a prospective study of 1,112 individuals (768 cognitively normal (CN), 133 with mild cognitive impairment (MCI), and 211 with Alzheimer’s disease dementia (AD)) as an ‘Inception cohort’ who underwent detailed ssessments every 18 months. Over the past decade, an additional 1247 subjects have been added as an ‘Enrichment cohort’ (as of 10 April 2019). Objective: Here we provide an overview of these Inception and Enrichment cohorts of more than 8,500 person-years of investigation. Methods: Participants underwent reassessment every 18 months including comprehensive cognitive testing, neuroimaging (magnetic resonance imaging, MRI; positron emission tomography, PET), biofluid biomarkers and lifestyle evaluations. Results: AIBL has made major contributions to the understanding of the natural history of AD, with cognitive and biological definitions of its three major stages: preclinical, prodromal and clinical. Early deployment of Aβ-amyloid and tau molecular PET imaging and the development of more sensitive and specific blood tests have facilitated the assessment of genetic and environmental factors which affect age at onset and rates of progression. Conclusion: This fifteen-year study provides a large database of highly characterized individuals with longitudinal cognitive, imaging and lifestyle data and biofluid collections, to aid in the development of interventions to delay onset, prevent or treat AD. Harmonization with similar large longitudinal cohort studies is underway to further these aims.
Collapse
Affiliation(s)
- Christopher Fowler
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,School of Psychological Science, University of Western Australia, Crawley, WA, Australia
| | - Sabine Bird
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Julia Bomke
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Pierrick Bourgeat
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Belinda M Brown
- Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Samantha C Burnham
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Ashley I Bush
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Carolyn Chadunow
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Steven Collins
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - James Doecke
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia.,Cooperative Research Council for Mental Health, Melbourne, VIC, Australia
| | - Vincent Doré
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia.,Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Kathryn A Ellis
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.,University of Melbourne Academic Unit for Psychiatry of Old Age, Parkville, VIC, Australia.,Melbourne School of Psychological Sciences, Melbourne, VIC, Australia
| | - Lis Evered
- Department of Anaesthesia and Acute Pain Medicine, St Vincent's Hospital Melbourne, Victoria Parade, Fitzroy, VIC, Australia
| | - Amir Fazlollahi
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Jurgen Fripp
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Samantha L Gardener
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Simon Gibson
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Robert Grenfell
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Elise Harrison
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Richard Head
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Liang Jin
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Adrian Kamer
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Fiona Lamb
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | | | - Simon M Laws
- Collaborative Genomics and Translation Group, Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Qiao-Xin Li
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Lucy Lim
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Yen Ying Lim
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.,Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Andrea Louey
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - S Lance Macaulay
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Lucy Mackintosh
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
| | | | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Simon McBride
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Lidija Milicic
- Collaborative Genomics and Translation Group, Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Madeline Peretti
- Collaborative Genomics and Translation Group, Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Kelly Pertile
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Tenielle Porter
- Collaborative Genomics and Translation Group, Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Morgan Radler
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Alan Rembach
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Joanne Robertson
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Mark Rodrigues
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Rebecca Rumble
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | | | - Greg Savage
- Department of Psychology, Macquarie University, Sydney, NSW, Australia
| | - Brendan Silbert
- Department of Anaesthesia and Acute Pain Medicine, St Vincent's Hospital Melbourne, Victoria Parade, Fitzroy, VIC, Australia
| | - Magdalene Soh
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Department of Biomedical Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Tania Taddei
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia
| | - Christine Thai
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Brett Trounson
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Regan Tyrrell
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Michael Vacher
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - Shiji Varghese
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Weinborn
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,School of Psychological Science, University of Western Australia, Crawley, WA, Australia
| | - Michael Woodward
- Department of Geriatric Medicine Austin Hospital, Heidelberg, VIC, Australia
| | - Ying Xia
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD, Australia
| | - David Ames
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.,University of Melbourne Academic Unit for Psychiatry of Old Age, Parkville, VIC, Australia.,National Ageing Research Institute (NARI), Parkville, VIC, Australia
| | | |
Collapse
|
28
|
Liu J, Yang W, Luo H, Ma Y, Zhao H, Dan X. Brain-derived neurotrophic factor Val66Met polymorphism is associated with mild cognitive impairment in elderly patients with type 2 diabetes: a case-controlled study. Aging Clin Exp Res 2021; 33:1659-1666. [PMID: 32892314 DOI: 10.1007/s40520-020-01687-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) Val66Met polymorphism is reported to be associated with cognitive dysfunction, an important comorbidity factor in patients with type 2 diabetes mellitus (T2DM), especially in elderly populations, however, the underlying pathophysiological mechanisms are unclear. AIM This study was performed to investigate the association between BDNF Val66Met polymorphism and mild cognitive impairment (MCI) in elderly patients with T2DM. METHODS In total, 105 MCI and 105 normal cognition controls of T2DM patients were enrolled; all of the patients underwent neuropsychological assessments. BDNF Val66Met polymorphism was genotyped via TaqMan SNP genotyping assay. Data from clinical and laboratory-based examinations were collected. RESULTS The frequency of the BDNF Met allele was significantly higher in the MCI group than in the controls. Multiple regression analysis indicated an association of the Met allele with MCI in patients with T2DM (OR = 2.54; 95% CI 1.33-4.84; p = 0.005). Stratified by educational level, the BDNF Met allele was significantly associated with MCI in elderly T2DM patients (OR = 3.29; 95% CI 1.26-8.57; p = 0.015) among the group of low educational levels (< 12 years); however, the association was insignificant among those with higher educational levels. DISCUSSION BDNF Met allele carriers showed a higher frequency of MCI than Val/Val homozygotes in elderly T2DM patients. However, this association was only significant in patients with low education levels. CONCLUSION BDNF Val66Met polymorphism may have a potential role in MCI in elderly T2DM patients, especially those with low educational levels.
Collapse
Affiliation(s)
- Jia Liu
- Department of Geriatric Medicine, Xuanwu Hospital, The Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Wei Yang
- Department of Geriatric Medicine, Xuanwu Hospital, The Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China.
| | - Hongyu Luo
- Department of Geriatric Medicine, Xuanwu Hospital, The Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Yixin Ma
- Department of Geriatric Medicine, Xuanwu Hospital, The Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Huan Zhao
- Department of Geriatric Medicine, Xuanwu Hospital, The Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Xiaojuan Dan
- Department of Neurology, Xuanwu Hospital, The Capital Medical University, Beijing, China
| |
Collapse
|
29
|
Jia J, Xu J, Liu J, Wang Y, Wang Y, Cao Y, Guo Q, Qu Q, Wei C, Wei W, Zhang J, Yu E. Comprehensive Management of Daily Living Activities, behavioral and Psychological Symptoms, and Cognitive Function in Patients with Alzheimer's Disease: A Chinese Consensus on the Comprehensive Management of Alzheimer's Disease. Neurosci Bull 2021; 37:1025-1038. [PMID: 34050523 PMCID: PMC8275730 DOI: 10.1007/s12264-021-00701-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/06/2021] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cognitive disorder in the elderly. Its main clinical manifestations are cognitive decline (C), behavioral and psychological symptoms (B), and a decline in the activities of daily living (A), also known as ABC symptoms. Early identification and evaluation of ABC symptoms are helpful for establishing the accurate diagnosis, comprehensive treatment, and prognosis of AD. To guide Chinese clinical practice for optimization of the comprehensive management of AD, in 2018, The Academy of Cognitive Disorder of China gathered 22 neurologists and gerontologists in China to build a consensus on the comprehensive management of AD. Based on a review of the evidence, the consensus summarizes the pathogenesis, pathological changes, clinical manifestations, evaluation, diagnosis, drug and non-drug treatment, and patient care for AD. Focus group discussion was used to establish a flowchart of comprehensive ABC management for AD patients. The new consensus provides a feasible AD management process for clinicians.
Collapse
Affiliation(s)
- Jianjun Jia
- Department of Neurology, The Second Medical Center, People's Liberation Army General Hospital, Beijing, 100853, China.
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Jun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yongjun Wang
- Cognitive Impairment Department, Shenzhen Kangning Hospital, Shenzhen, 518118, China
| | - Yanjiang Wang
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yunpeng Cao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, 210112, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Qiuming Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Cuibai Wei
- Department of Neurology, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Wenshi Wei
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Enyan Yu
- Department of Psychology, Chinese Academy of Sciences Cancer Hospital of the University of the Chinese Academy of Sciences, Hangzhou, 310022, China
| |
Collapse
|
30
|
Lee B, Shin M, Park Y, Won SY, Cho KS. Physical Exercise-Induced Myokines in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22115795. [PMID: 34071457 PMCID: PMC8198301 DOI: 10.3390/ijms22115795] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS), are disorders characterized by progressive degeneration of the nervous system. Currently, there is no disease-modifying treatments for most NDs. Meanwhile, numerous studies conducted on human and animal models over the past decades have showed that exercises had beneficial effects on NDs. Inter-tissue communication by myokine, a peptide produced and secreted by skeletal muscles during exercise, is thought to be an important underlying mechanism for the advantages. Here, we reviewed studies about the effects of myokines regulated by exercise on NDs and their mechanisms. Myokines could exert beneficial effects on NDs through a variety of regulatory mechanisms, including cell survival, neurogenesis, neuroinflammation, proteostasis, oxidative stress, and protein modification. Studies on exercise-induced myokines are expected to provide a novel strategy for treating NDs, for which there are no adequate treatments nowadays. To date, only a few myokines have been investigated for their effects on NDs and studies on mechanisms involved in them are in their infancy. Therefore, future studies are needed to discover more myokines and test their effects on NDs.
Collapse
Affiliation(s)
- Banseok Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - Myeongcheol Shin
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - Youngjae Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - So-Yoon Won
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
- Korea Hemp Institute, Konkuk University, Seoul 05029, Korea
- Correspondence: (S.-Y.W.); (K.S.C.); Tel.: +82-10-3688-5474 (S.-Y.W.); Tel.: +82-2-450-3424 (K.S.C.)
| | - Kyoung Sang Cho
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
- Korea Hemp Institute, Konkuk University, Seoul 05029, Korea
- Correspondence: (S.-Y.W.); (K.S.C.); Tel.: +82-10-3688-5474 (S.-Y.W.); Tel.: +82-2-450-3424 (K.S.C.)
| |
Collapse
|
31
|
Cechova K, Andel R, Angelucci F, Chmatalova Z, Markova H, Laczó J, Vyhnalek M, Matoska V, Kaplan V, Nedelska Z, Ward DD, Hort J. Impact of APOE and BDNF Val66Met Gene Polymorphisms on Cognitive Functions in Patients with Amnestic Mild Cognitive Impairment. J Alzheimers Dis 2021; 73:247-257. [PMID: 31771052 DOI: 10.3233/jad-190464] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Apolipoprotein (APOE) ɛ4 is a well-known risk factor for late-onset Alzheimer's disease (AD), but other AD-related gene polymorphisms might also be important, such as the polymorphism within the brain-derived neurotrophic factor (BDNF) gene. Carriage of BDNF Val66Met has been associated with faster cognitive decline and greater hippocampal atrophy in cognitively normal elderly. Thus, we examined the effects of the concurrent presence of APOE and BDNF polymorphisms on cognitive functions and brain morphometry in amnestic mild cognitive impairment (aMCI) patients. 107 aMCI patients (mean age = 72.2) were recruited from the Czech Brain Aging Study and, based on APOE and BDNF genes polymorphisms, were divided into four groups: ɛ4-BDNFVal/Val (n = 37), ɛ4-BDNFMet (n = 19), ɛ4+BDNFVal/Val (n = 35), and ɛ4+BDNFMet (n = 16). All patients underwent clinical examination, magnetic resonance imaging, and complex neuropsychological battery. The combination of APOEɛ4+ and BDNF Met was associated with significantly worse memory performance in immediate and delayed recall compared to other polymorphism groups. We did not observe increased atrophy in areas related to memory function in the ɛ4+BDNFMet group. Our findings suggest that carriage of ɛ4+BDNFMet is associated with more pronounced memory dysfunction, a typical feature of early AD, but not with structural brain changes in aMCI patients. These findings suggest that in APOEɛ4/BDNF Met carriers, synaptic dysfunction affecting memory may precede pronounced structural changes.
Collapse
Affiliation(s)
- Katerina Cechova
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Ross Andel
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,School of Aging Studies, University of South Florida, Tampa, FL, USA
| | - Francesco Angelucci
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Zuzana Chmatalova
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hana Markova
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jan Laczó
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Martin Vyhnalek
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Vaclav Matoska
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Vojtech Kaplan
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Zuzana Nedelska
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - David D Ward
- Department of Medicine, Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,Centre for Health Care of the Elderly, QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Jakub Hort
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
32
|
Mentis AFA, Dardiotis E, Chrousos GP. Apolipoprotein E4 and meningeal lymphatics in Alzheimer disease: a conceptual framework. Mol Psychiatry 2021; 26:1075-1097. [PMID: 32355332 PMCID: PMC7985019 DOI: 10.1038/s41380-020-0731-7] [Citation(s) in RCA: 48] [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: 09/03/2019] [Revised: 04/01/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
The potential existence and roles of the meningeal lymphatic system in normal and pathological brain function have been a long-standing enigma. Recent evidence suggests that meningeal lymphatic vessels are present in both the mouse and human brain; in mice, they seem to play a role in clearing toxic amyloid-beta peptides, which have been connected with Alzheimer disease (AD). Here, we review the evidence linking the meningeal lymphatic system with human AD. Novel findings suggest that the recently described meningeal lymphatic vessels could be linked to, and possibly drain, the efferent paravascular glial lymphatic (glymphatic) system carrying cerebrospinal fluid, after solute and immune cell exchange with brain interstitial fluid. In so doing, the glymphatic system could contribute to the export of toxic solutes and immune cells from the brain (an exported fluid we wish to describe as glymph, similarly to lymph) to the meningeal lymphatic system; the latter, by being connected with downstream anatomic regions, carries the glymph to the conventional cervical lymphatic vessels and nodes. Thus, abnormal function in the meningeal lymphatic system could, in theory, lead to the accumulation, in the brain, of amyloid-beta, cellular debris, and inflammatory mediators, as well as immune cells, resulting in damage of the brain parenchyma and, in turn, cognitive and other neurologic dysfunctions. In addition, we provide novel insights into APOE4-the leading genetic risk factor for AD-and its relation to the meningeal lymphatic system. In this regard, we have reanalyzed previously published RNA-Seq data to show that induced pluripotent stem cells (iPSCs) carrying the APOE4 allele (either as APOE4 knock-in or stemming from APOE4 patients) express lower levels of (a) genes associated with lymphatic markers, and (b) genes for which well-characterized missense mutations have been linked to peripheral lymphedema. Taking into account this evidence, we propose a new conceptual framework, according to which APOE4 could play a novel role in the premature shrinkage of meningeal lymphatic vessels (meningeal lymphosclerosis), leading to abnormal meningeal lymphatic functions (meningeal lymphedema), and, in turn, reduction in the clearance of amyloid-beta and other macromolecules and inflammatory mediators, as well as immune cells, from the brain, exacerbation of AD manifestations, and progression of the disease. Altogether, these findings and their potential interpretations may herald novel diagnostic tools and therapeutic approaches in patients with AD.
Collapse
Affiliation(s)
- Alexios-Fotios A Mentis
- Public Health Laboratories, Hellenic Pasteur Institute, Vas. Sofias Avenue 127, 115 21, Athens, Greece.
- Department of Microbiology, University of Thessaly, Panepistimiou 3, Viopolis, 41 500, Larissa, Greece.
| | - Efthimios Dardiotis
- Department of Neurology, University of Thessaly, Panepistimiou 3, Viopolis, 41 500, Larissa, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Medical School, Aghia Sophia Children's Hospital, Livadias 8, 115 27, Athens, Greece
- UNESCO Chair on Adolescent Health Care, Athens, Greece
| |
Collapse
|
33
|
Fernandez S, Burnham SC, Milicic L, Savage G, Maruff P, Peretti M, Sohrabi HR, Lim YY, Weinborn M, Ames D, Masters CL, Martins RN, Rainey-Smith S, Rowe CC, Salvado O, Groth D, Verdile G, Villemagne VL, Porter T, Laws SM. SPON1 Is Associated with Amyloid-β and APOE ε4-Related Cognitive Decline in Cognitively Normal Adults. J Alzheimers Dis Rep 2021; 5:111-120. [PMID: 33782664 PMCID: PMC7990462 DOI: 10.3233/adr-200246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract.
Collapse
Affiliation(s)
- Shane Fernandez
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Samantha C Burnham
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,CSIRO Health and Biosecurity, Parkville, Victoria, Australia
| | - Lidija Milicic
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Greg Savage
- ARC Centre of Excellence in Cognition and its Disorders, Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.,CogState Ltd., Melbourne, Victoria, Australia
| | - Madeline Peretti
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre for Healthy Ageing, Murdoch University, Murdoch, Western Australia, Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Yen Ying Lim
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Michael Weinborn
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, St. Vincent's Health, The University of Melbourne, Kew, Victoria, Australia.,National Ageing Research Institute, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph N Martins
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Stephanie Rainey-Smith
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Olivier Salvado
- CSIRO Health and Biosecurity/Australian e-Health Research Centre, Herston, Queensland, Australia
| | - David Groth
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Giuseppe Verdile
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Victor L Villemagne
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Tenielle Porter
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Simon M Laws
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| |
Collapse
|
34
|
Brown BM, Frost N, Rainey-Smith SR, Doecke J, Markovic S, Gordon N, Weinborn M, Sohrabi HR, Laws SM, Martins RN, Erickson KI, Peiffer JJ. High-intensity exercise and cognitive function in cognitively normal older adults: a pilot randomised clinical trial. Alzheimers Res Ther 2021; 13:33. [PMID: 33522961 PMCID: PMC7849126 DOI: 10.1186/s13195-021-00774-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/15/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND Physical inactivity has been consistently linked to increased risk of cognitive decline; however, studies examining the impact of exercise interventions on cognition have produced inconsistent findings. Some observational studies suggest exercise intensity may be important for inducing cognitive improvements; however, this has yet to be thoroughly examined in older adult cohorts. The objective of the current study was to evaluate the effect of systematically manipulated high-intensity and moderate-intensity exercise interventions on cognition. METHODS This multi-arm pilot randomised clinical trial investigated the effects of 6 months of high-intensity exercise and moderate-intensity exercise, compared with an inactive control, on cognition. Outcome measures were assessed at pre- (baseline), post- (6 months), and 12 months post-intervention. Ninety-nine cognitively normal men and women (aged 60-80 years) were enrolled from October 2016 to November 2017. Participants that were allocated to an exercise group (i.e. high-intensity or moderate-intensity) engaged in cycle-based exercise two times per week for 6 months. Cognition was assessed using a comprehensive neuropsychological test battery. Cardiorespiratory fitness was evaluated by a graded exercise test. RESULTS There was a dose-dependent effect of exercise intensity on cardiorespiratory fitness, whereby the high-intensity group experienced greater increases in fitness than the moderate-intensity and control groups. However, there was no direct effect of exercise on cognition. CONCLUSIONS We did not observe a direct effect of exercise on cognition. Future work in this field should be appropriately designed and powered to examine factors that may contribute to individual variability in response to intervention. TRIAL REGISTRATION This study is registered with the Australian New Zealand Clinical Trials Registry (ACTRN12617000643370). Registered on 3 May 2017-retrospectively registered. https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=372780.
Collapse
Affiliation(s)
- Belinda M Brown
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, 6150, Australia.
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia.
- Centre for Healthy Ageing, Murdoch University, Murdoch, Western Australia, Australia.
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.
| | - Natalie Frost
- School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - Stephanie R Rainey-Smith
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
- Centre for Healthy Ageing, Murdoch University, Murdoch, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - James Doecke
- Australian eHealth Research Centre, CSIRO, Brisbane, Queensland, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Shaun Markovic
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, 6150, Australia
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Nicole Gordon
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, 6150, Australia
| | - Michael Weinborn
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
- School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
- Centre for Healthy Ageing, Murdoch University, Murdoch, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Simon M Laws
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Ralph N Martins
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Kirk I Erickson
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, 6150, Australia
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeremiah J Peiffer
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, 6150, Australia
- Centre for Healthy Ageing, Murdoch University, Murdoch, Western Australia, Australia
| |
Collapse
|
35
|
Daily oscillation of cognitive factors is modified in the temporal cortex of an amyloid β(1-42)-induced rat model of Alzheimer's disease. Brain Res Bull 2021; 170:106-114. [PMID: 33508401 DOI: 10.1016/j.brainresbull.2021.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/28/2020] [Accepted: 01/20/2021] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD) is a devastating disease characterized by loss of synapses and neurons in the elderly. Accumulation of the β-amyloid peptide (Aβ) in the brain is thought to be central to the pathogenesis of AD. ApoE plays a key role in normal and physiological clearance of Aß, since it facilitates the peptide intra- and extracellular proteolytic degradation. Besides the cognitive deficit, AD patients also show alterations in their circadian rhythms. The objective of this study was to investigate the effects of an i.c.v. injection of Aβ (1-42) peptide on the 24 h rhythms of Apo E, BMAL1, RORα, Bdnf and trkB mRNA and Aβ levels in the rat temporal cortex. We found that an i.c.v. injection of Aβ aggregates phase shifts daily Bdnf expression as well as Apo E, BMAL1, RORα, Aβ and decreased the mesor of TrkB rhythms. Thus, elevated Aβ peptide levels might modify the temporal patterns of cognition-related factors, probably; by affecting the clock factors rhythms as well as in the 24 h rhythms of Apo E.
Collapse
|
36
|
Jung M, Apostolova LG, Gao S, Burney HN, Lai D, Foroud T, Saykin AJ, Pressler SJ. Testing influences of APOE and BDNF genes and heart failure on cognitive function. Heart Lung 2021; 50:51-58. [PMID: 32703621 PMCID: PMC8809626 DOI: 10.1016/j.hrtlng.2020.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Apolipoprotein E (APOE) ε2, ε4 and brain-derived neurotrophic factor (BDNF) Val66Met alleles have been associated with cognition. Associations of these alleles with cognition in heart failure (HF) and influences of HF across the cognitive spectrum (i.e., cognitively normal to Alzheimer's dementia [AD]) remain unexplored. OBJECTIVES To investigate influences of APOE ε2, ε4, BDNF Met and HF on cognition among participants across the cognitive spectrum. METHODS Genetic association study using national databases (N = 7,166). RESULTS APOE ε2 frequencies were similar across the cognitive spectrum among participants with HF. APOE ε4 frequency was lower among participants with HF and AD than non-HF participants with AD. BDNF Met frequencies did not differ across the spectrum. HF was associated with worse attention and language. In the HF subsample, ε4 was associated with worse memory. CONCLUSION Associations between APOE and cognition may differ in HF but need to be tested in a larger sample.
Collapse
Affiliation(s)
- Miyeon Jung
- Assistant Professor, Indiana University School of Nursing, 600 Barnhill Drive, Indianapolis, IN 46202, USA.
| | - Liana G Apostolova
- Professor, Indiana University School of Medicine, Neurology, Radiology, Medical and Molecular Genetics, 355 West 16th Street, Indianapolis, IN 46202, USA.
| | - Sujuan Gao
- Professor, Indiana University School of Medicine, Department of Biostatistics, 410 West 10th Street, Indianapolis, IN 46202, USA.
| | - Heather N Burney
- Biostatistician, Indiana University School of Medicine, Department of Biostatistics, 410 West 10th Street, Indianapolis, IN 46202, USA.
| | - Dongbing Lai
- Assistant Research Professor, Indiana University School of Medicine, Medical and Molecular Genetics, 410 West 10th Street, Indianapolis, IN 46202, USA.
| | - Tatiana Foroud
- Professor, Indiana University School of Medicine, Medical and Molecular Genetics, 410 West 10th Street, Indianapolis, IN 46202, USA.
| | - Andrew J Saykin
- Professor, Indiana University School of Medicine, Department of Radiology and Imaging Sciences, 355 West 16th street, Indianapolis, IN 46202, USA.
| | - Susan J Pressler
- Professor, Indiana University School of Nursing, 600 Barnhill Drive, Indianapolis, IN 46202, USA.
| |
Collapse
|
37
|
Franzmeier N, Ren J, Damm A, Monté-Rubio G, Boada M, Ruiz A, Ramirez A, Jessen F, Düzel E, Rodríguez Gómez O, Benzinger T, Goate A, Karch CM, Fagan AM, McDade E, Buerger K, Levin J, Duering M, Dichgans M, Suárez-Calvet M, Haass C, Gordon BA, Lim YY, Masters CL, Janowitz D, Catak C, Wolfsgruber S, Wagner M, Milz E, Moreno-Grau S, Teipel S, Grothe MJ, Kilimann I, Rossor M, Fox N, Laske C, Chhatwal J, Falkai P, Perneczky R, Lee JH, Spottke A, Boecker H, Brosseron F, Fliessbach K, Heneka MT, Nestor P, Peters O, Fuentes M, Menne F, Priller J, Spruth EJ, Franke C, Schneider A, Westerteicher C, Speck O, Wiltfang J, Bartels C, Araque Caballero MÁ, Metzger C, Bittner D, Salloway S, Danek A, Hassenstab J, Yakushev I, Schofield PR, Morris JC, Bateman RJ, Ewers M. The BDNF Val66Met SNP modulates the association between beta-amyloid and hippocampal disconnection in Alzheimer's disease. Mol Psychiatry 2021; 26:614-628. [PMID: 30899092 PMCID: PMC6754794 DOI: 10.1038/s41380-019-0404-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/19/2019] [Accepted: 02/14/2019] [Indexed: 01/29/2023]
Abstract
In Alzheimer's disease (AD), a single-nucleotide polymorphism in the gene encoding brain-derived neurotrophic factor (BDNFVal66Met) is associated with worse impact of primary AD pathology (beta-amyloid, Aβ) on neurodegeneration and cognitive decline, rendering BDNFVal66Met an important modulating factor of cognitive impairment in AD. However, the effect of BDNFVal66Met on functional networks that may underlie cognitive impairment in AD is poorly understood. Using a cross-validation approach, we first explored in subjects with autosomal dominant AD (ADAD) from the Dominantly Inherited Alzheimer Network (DIAN) the effect of BDNFVal66Met on resting-state fMRI assessed functional networks. In seed-based connectivity analysis of six major large-scale networks, we found a stronger decrease of hippocampus (seed) to medial-frontal connectivity in the BDNFVal66Met carriers compared to BDNFVal homozogytes. BDNFVal66Met was not associated with connectivity in any other networks. Next, we tested whether the finding of more pronounced decrease in hippocampal-medial-frontal connectivity in BDNFVal66Met could be also found in elderly subjects with sporadically occurring Aβ, including a group with subjective cognitive decline (N = 149, FACEHBI study) and a group ranging from preclinical to AD dementia (N = 114, DELCODE study). In both of these independently recruited groups, BDNFVal66Met was associated with a stronger effect of more abnormal Aβ-levels (assessed by biofluid-assay or amyloid-PET) on hippocampal-medial-frontal connectivity decreases, controlled for hippocampus volume and other confounds. Lower hippocampal-medial-frontal connectivity was associated with lower global cognitive performance in the DIAN and DELCODE studies. Together these results suggest that BDNFVal66Met is selectively associated with a higher vulnerability of hippocampus-frontal connectivity to primary AD pathology, resulting in greater AD-related cognitive impairment.
Collapse
Affiliation(s)
- Nicolai Franzmeier
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Jinyi Ren
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Alexander Damm
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Gemma Monté-Rubio
- grid.477255.60000 0004 1765 5601Fundació ACE, Alzheimer Treatment and Research Center, Barcelona, Spain
| | - Mercè Boada
- grid.477255.60000 0004 1765 5601Fundació ACE, Alzheimer Treatment and Research Center, Barcelona, Spain ,grid.451322.30000 0004 1770 9462CIBERNED, Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Agustín Ruiz
- grid.477255.60000 0004 1765 5601Fundació ACE, Alzheimer Treatment and Research Center, Barcelona, Spain ,grid.451322.30000 0004 1770 9462CIBERNED, Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Alfredo Ramirez
- grid.6190.e0000 0000 8580 3777Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany ,grid.10388.320000 0001 2240 3300Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Frank Jessen
- grid.6190.e0000 0000 8580 3777Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Emrah Düzel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Octavio Rodríguez Gómez
- grid.477255.60000 0004 1765 5601Fundació ACE, Alzheimer Treatment and Research Center, Barcelona, Spain ,grid.451322.30000 0004 1770 9462CIBERNED, Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Tammie Benzinger
- grid.4367.60000 0001 2355 7002Department of Radiology, Washington University in St Louis, St Louis, MO USA ,grid.4367.60000 0001 2355 7002Knight Alzheimer’s Disease Research Center, Washington University in St. Louis, St. Louis, MO USA
| | - Alison Goate
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Ronald M. Loeb Center for Alzheimer’s Disease, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Celeste M. Karch
- grid.4367.60000 0001 2355 7002Knight Alzheimer’s Disease Research Center, Washington University in St. Louis, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University in St Louis, St Louis, MO USA
| | - Anne M. Fagan
- grid.4367.60000 0001 2355 7002Knight Alzheimer’s Disease Research Center, Washington University in St. Louis, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Department of Neurology, Washington University in St. Louis, St. Louis, MO USA
| | - Eric McDade
- grid.4367.60000 0001 2355 7002Department of Neurology, Washington University in St. Louis, St. Louis, MO USA
| | - Katharina Buerger
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Johannes Levin
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.5252.00000 0004 1936 973XDepartment of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marco Duering
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Martin Dichgans
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marc Suárez-Calvet
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.430077.7Barcelonabeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Catalonia Spain ,grid.5252.00000 0004 1936 973XFaculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Haass
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.5252.00000 0004 1936 973XFaculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Brian A. Gordon
- grid.4367.60000 0001 2355 7002Knight Alzheimer’s Disease Research Center, Washington University in St. Louis, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Department of Psychological and Brain Sciences, Washington University, St. Louis, MO USA
| | - Yen Ying Lim
- grid.1008.90000 0001 2179 088XThe Florey Institute, The University of Melbourne, Parkville, VIC Australia
| | - Colin L. Masters
- grid.1008.90000 0001 2179 088XThe Florey Institute, The University of Melbourne, Parkville, VIC Australia
| | - Daniel Janowitz
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Cihan Catak
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Steffen Wolfsgruber
- grid.10388.320000 0001 2240 3300Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael Wagner
- grid.10388.320000 0001 2240 3300Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Esther Milz
- grid.6190.e0000 0000 8580 3777Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Sonia Moreno-Grau
- grid.477255.60000 0004 1765 5601Fundació ACE, Alzheimer Treatment and Research Center, Barcelona, Spain ,grid.451322.30000 0004 1770 9462CIBERNED, Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Stefan Teipel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany ,grid.413108.f0000 0000 9737 0454Department of Psychosomatic Medicine, University Hospital Rostock, Rostock, Germany
| | - Michel J Grothe
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Ingo Kilimann
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Martin Rossor
- grid.83440.3b0000000121901201Dementia Research Centre, University College London, Queen Square, London, UK
| | - Nick Fox
- grid.83440.3b0000000121901201Dementia Research Centre, University College London, Queen Square, London, UK
| | - Christoph Laske
- grid.428620.aHertie Institute for Clinical Brain Research, Tübingen, Germany ,grid.424247.30000 0004 0438 0426Germany and German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Jasmeer Chhatwal
- grid.38142.3c000000041936754XMassachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston, MA USA
| | - Peter Falkai
- grid.5252.00000 0004 1936 973XDepartment of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Robert Perneczky
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany ,grid.5252.00000 0004 1936 973XDepartment of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, Munich, Germany ,grid.7445.20000 0001 2113 8111Neuroepidemiology and Ageing Research Unit, School of Public Health, The Imperial College of Science, Technology and Medicine, London, UK
| | - Jae-Hong Lee
- grid.413967.e0000 0001 0842 2126Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Annika Spottke
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany ,grid.10388.320000 0001 2240 3300Department of Neurology, University of Bonn, Bonn, Germany
| | - Henning Boecker
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany ,grid.10388.320000 0001 2240 3300Department of Radiology, University of Bonn, Bonn, Germany
| | - Frederic Brosseron
- grid.10388.320000 0001 2240 3300Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Klaus Fliessbach
- grid.10388.320000 0001 2240 3300Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T. Heneka
- grid.10388.320000 0001 2240 3300Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Peter Nestor
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany ,grid.1003.20000 0000 9320 7537Queensland Brain Institute, University of Queensland, Brisbane, QLD Australia
| | - Oliver Peters
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Manuel Fuentes
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Felix Menne
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Josef Priller
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Neuropsychiatry, Charité, Berlin, Germany
| | - Eike J. Spruth
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Neuropsychiatry, Charité, Berlin, Germany
| | - Christiana Franke
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Neuropsychiatry, Charité, Berlin, Germany
| | - Anja Schneider
- grid.10388.320000 0001 2240 3300Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Christine Westerteicher
- grid.10388.320000 0001 2240 3300Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Oliver Speck
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany ,grid.418723.b0000 0001 2109 6265Leibniz Institute for Neurobiology, Magdeburg, Germany ,grid.452320.20000 0004 0404 7236Center for Behavioral Brain Sciences, Magdeburg, Germany ,grid.5807.a0000 0001 1018 4307Department for Biomedical Magnetic Resonance, Institute for Physics, Otto-von-Guericke University, Magdeburg, Germany
| | - Jens Wiltfang
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany ,grid.7450.60000 0001 2364 4210Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany ,grid.7311.40000000123236065iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Claudia Bartels
- grid.7450.60000 0001 2364 4210Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
| | - Miguel Ángel Araque Caballero
- grid.5252.00000 0004 1936 973XInstitute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Coraline Metzger
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Daniel Bittner
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Stephen Salloway
- grid.40263.330000 0004 1936 9094Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI USA
| | - Adrian Danek
- grid.5252.00000 0004 1936 973XDepartment of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jason Hassenstab
- grid.4367.60000 0001 2355 7002Department of Neurology, Washington University in St. Louis, St. Louis, MO USA
| | - Igor Yakushev
- grid.6936.a0000000123222966Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Peter R. Schofield
- grid.250407.40000 0000 8900 8842Neuroscience Research Australia, Barker Street Randwick, Sydney, NSW 2031 Australia ,grid.1005.40000 0004 4902 0432School of Medical Sciences, University of New South Wales, Sydney, NSW 2052 Australia
| | - John C. Morris
- grid.4367.60000 0001 2355 7002Knight Alzheimer’s Disease Research Center, Washington University in St. Louis, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University in St Louis, St Louis, MO USA ,grid.4367.60000 0001 2355 7002Department of Neurology, Washington University in St. Louis, St. Louis, MO USA
| | - Randall J. Bateman
- grid.4367.60000 0001 2355 7002Knight Alzheimer’s Disease Research Center, Washington University in St. Louis, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Department of Neurology, Washington University in St. Louis, St. Louis, MO USA
| | - Michael Ewers
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany.
| |
Collapse
|
38
|
Lim YY, Laws SM, Perin S, Pietrzak RH, Fowler C, Masters CL, Maruff P. BDNF VAL66MET polymorphism and memory decline across the spectrum of Alzheimer's disease. GENES BRAIN AND BEHAVIOR 2020; 20:e12724. [PMID: 33369083 DOI: 10.1111/gbb.12724] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/07/2020] [Accepted: 12/20/2020] [Indexed: 12/29/2022]
Abstract
The brain-derived neurotrophic factor (BDNF) Val66Met (rs6265) polymorphism has been shown to moderate the extent to which memory decline manifests in preclinical Alzheimer's disease (AD). To date, no study has examined the relationship between BDNF and memory in individuals across biologically confirmed AD clinical stages (i.e., Aβ+). We aimed to understand the effect of BDNF on episodic memory decline and clinical disease progression over 126 months in individuals with preclinical, prodromal and clinical AD. Participants enrolled in the Australian Imaging, Biomarkers and Lifestyle (AIBL) study who were Aβ + (according to positron emission tomography), and cognitively normal (CN; n = 238), classified as having mild cognitive impairment (MCI; n = 80), or AD (n = 66) were included in this study. Cognition was evaluated at 18 month intervals using an established episodic memory composite score over 126 months. We observed that in Aβ + CNs, Met66 was associated with greater memory decline with increasing age and were 1.5 times more likely to progress to MCI/AD over 126 months. In Aβ + MCIs, there was no effect of Met66 on memory decline or on disease progression to AD over 126 months. In Aβ + AD, Val66 homozygotes showed greater memory decline, while Met66 carriers performed at a constant and very impaired level. Our current results illustrate the importance of time and disease severity to clinicopathological models of the role of BDNF Val66Met in memory decline and AD clinical progression. Specifically, the effect of BDNF on memory decline is greatest in preclinical AD and reduces as AD clinical disease severity increases.
Collapse
Affiliation(s)
- Yen Ying Lim
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Simon M Laws
- Collaborative Genomics and Translation Group, Strategic Research Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Stephanie Perin
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Robert H Pietrzak
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Christopher Fowler
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia.,Cogstate Ltd, Melbourne, Australia
| | | |
Collapse
|
39
|
Manning KJ, Steffens DC. Blue Genes, Exercise, and Cognition in Late-Life Depression: A Lot of Moving Parts. Am J Geriatr Psychiatry 2020; 28:968-970. [PMID: 32654839 DOI: 10.1016/j.jagp.2020.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Kevin J Manning
- Department of Psychiatry (KJM, DCS), University of Connecticut Health Center, Farmington, CT.
| | - David C Steffens
- Department of Psychiatry (KJM, DCS), University of Connecticut Health Center, Farmington, CT
| |
Collapse
|
40
|
Vilor-Tejedor N, Operto G, Evans TE, Falcon C, Crous-Bou M, Minguillón C, Cacciaglia R, Milà-Alomà M, Grau-Rivera O, Suárez-Calvet M, Garrido-Martín D, Morán S, Esteller M, Adams HH, Molinuevo JL, Guigó R, Gispert JD. Effect of BDNF Val66Met on hippocampal subfields volumes and compensatory interaction with APOE-ε4 in middle-age cognitively unimpaired individuals from the ALFA study. Brain Struct Funct 2020; 225:2331-2345. [PMID: 32804326 PMCID: PMC7544723 DOI: 10.1007/s00429-020-02125-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/30/2020] [Indexed: 11/08/2022]
Abstract
Background Current evidence supports the involvement of brain-derived neurotrophic factor (BDNF) Val66Met polymorphism, and the ε4 allele of APOE gene in hippocampal-dependent functions. Previous studies on the association of Val66Met with whole hippocampal volume included patients of a variety of disorders. However, it remains to be elucidated whether there is an impact of BDNF Val66Met polymorphism on the volumes of the hippocampal subfield volumes (HSv) in cognitively unimpaired (CU) individuals, and the interactive effect with the APOE-ε4 status. Methods BDNF Val66Met and APOE genotypes were determined in a sample of 430 CU late/middle-aged participants from the ALFA study (ALzheimer and FAmilies). Participants underwent a brain 3D-T1-weighted MRI scan, and volumes of the HSv were determined using Freesurfer (v6.0). The effects of the BDNF Val66Met genotype on the HSv were assessed using general linear models corrected by age, gender, education, number of APOE-ε4 alleles and total intracranial volume. We also investigated whether the association between APOE-ε4 allele and HSv were modified by BDNF Val66Met genotypes. Results BDNF Val66Met carriers showed larger bilateral volumes of the subiculum subfield. In addition, HSv reductions associated with APOE-ε4 allele were significantly moderated by BDNF Val66Met status. BDNF Met carriers who were also APOE-ε4 homozygous showed patterns of higher HSv than BDNF Val carriers. Conclusion To our knowledge, the present study is the first to show that carrying the BDNF Val66Met polymorphisms partially compensates the decreased on HSv associated with APOE-ε4 in middle-age cognitively unimpaired individuals. Electronic supplementary material The online version of this article (10.1007/s00429-020-02125-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Natalia Vilor-Tejedor
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, C. Doctor Aiguader 88, Edif. PRBB, 08003, Barcelona, Spain. .,Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain. .,Erasmus MC University Medical Center Rotterdam, Department of Clinical Genetics, Rotterdam, The Netherlands. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| | - Grégory Operto
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Tavia E Evans
- Erasmus MC University Medical Center Rotterdam, Department of Clinical Genetics, Rotterdam, The Netherlands
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Marta Crous-Bou
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain
| | - Carolina Minguillón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Diego Garrido-Martín
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, C. Doctor Aiguader 88, Edif. PRBB, 08003, Barcelona, Spain
| | - Sebastián Morán
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
| | - Hieab H Adams
- Erasmus MC University Medical Center Rotterdam, Department of Clinical Genetics, Rotterdam, The Netherlands.,Erasmus MC University Medical Center Rotterdam, Department of Epidemiology, Rotterdam, The Netherlands.,Erasmus MC University Medical Center Rotterdam, Department of Radiology, Rotterdam, The Netherlands
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, C. Doctor Aiguader 88, Edif. PRBB, 08003, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain. .,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.
| | | |
Collapse
|
41
|
Amidfar M, de Oliveira J, Kucharska E, Budni J, Kim YK. The role of CREB and BDNF in neurobiology and treatment of Alzheimer's disease. Life Sci 2020; 257:118020. [PMID: 32603820 DOI: 10.1016/j.lfs.2020.118020] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia worldwide. β-amyloid peptide (Aβ) is currently assumed to be the main cause of synaptic dysfunction and cognitive impairments in AD, but the molecular signaling pathways underlying its neurotoxic consequences have not yet been completely explored. Additional investigations regarding these pathways will contribute to development of new therapeutic targets. In context, developing evidence suggest that Aβ decreases brain-derived neurotrophic factor (BDNF) mostly by lowering phosphorylated cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) protein. In fact, it has been observed that brain or serum levels of BDNF appear to be beneficial markers for cognitive condition. In addition, the participation of transcription mediated by CREB has been widely analyzed in the memory process and AD development. Designing pharmacologic or genetic therapeutic approaches based on the targeting of CREB-BDNF signaling could be a promising treatment potential for AD. In this review, we summarize data demonstrating the role of CREB-BDNF signaling pathway in cognitive status and mediation of Aβ toxicity in AD. Finally, we also focus on the developing intervention methods for improvement of cognitive decline in AD based on targeting of CREB-BDNF pathway.
Collapse
Affiliation(s)
| | - Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ewa Kucharska
- Jesuit University Ignatianum in Krakow, Faculty of Education, Institute of Educational Sciences, Poland
| | - Josiane Budni
- Laboratório de Neurologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - Yong-Ku Kim
- Departments of Psychiatry, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
42
|
Bessi V, Mazzeo S, Bagnoli S, Padiglioni S, Carraro M, Piaceri I, Bracco L, Sorbi S, Nacmias B. The implication of BDNF Val66Met polymorphism in progression from subjective cognitive decline to mild cognitive impairment and Alzheimer's disease: a 9-year follow-up study. Eur Arch Psychiatry Clin Neurosci 2020; 270:471-482. [PMID: 31560105 DOI: 10.1007/s00406-019-01069-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
Abstract
Brain-derived natriuretic factor (BDNF) Val66Met polymorphism has been frequently reported to be associated with Alzheimer's disease (AD) with contrasting results. Numerous studies showed that Met allele increased the risk of AD only in women, while other studies have found worse cognitive performance in Val/Val carriers. We aimed to inquire the effects of Val66Met polymorphism on the progression from subjective cognitive decline (SCD) to mild cognitive impairment (MCI) and from MCI to AD and to ascertain if this effect is modulated by demographic and cognitive variables. For this purpose, we followed up 74 subjects (48 SCD, 26 MCI) for a mean time of 9 years. All participants underwent extensive neuropsychological assessment, cognitive reserve estimation, BDNF and apolipoprotein E (ApoE) genotype analysis at baseline. Personality traits and leisure activities were assessed in a subgroup. Each patient underwent clinical-neuropsychological follow-up, during which 18 out of 48 SCD subjects progressed to MCI and 14 out of 26 MCI subjects progressed to AD. We found that Val66Met increased the risk of progression from SCD to MCI and from MCI to AD only in women. Nevertheless, Val/Val carriers who progressed from SCD to MCI had a shorter conversion time compared to Met carriers. We concluded that Val66Met polymorphism might play different roles depending on sex and stage of the disease.
Collapse
Affiliation(s)
- Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy.
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Sonia Padiglioni
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Marco Carraro
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Laura Bracco
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| |
Collapse
|
43
|
Masters CL. Major risk factors for Alzheimer's disease: age and genetics. Lancet Neurol 2020; 19:475-476. [PMID: 32470411 DOI: 10.1016/s1474-4422(20)30155-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Colin L Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| |
Collapse
|
44
|
Wang S, Yao H, Xu Y, Hao R, Zhang W, Liu H, Huang Y, Guo W, Lu B. Therapeutic potential of a TrkB agonistic antibody for Alzheimer's disease. Theranostics 2020; 10:6854-6874. [PMID: 32550908 PMCID: PMC7295064 DOI: 10.7150/thno.44165] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022] Open
Abstract
Repeated failures of "Aβ-lowering" therapies call for new targets and therapeutic approaches for Alzheimer's disease (AD). We propose to treat AD by halting neuronal death and repairing synapses using a BDNF-based therapy. To overcome the poor druggability of BDNF, we have developed an agonistic antibody AS86 to mimic the function of BDNF, and evaluate its therapeutic potential for AD. Method: Biochemical, electrophysiological and behavioral techniques were used to investigate the effects of AS86 in vitro and in vivo. Results: AS86 specifically activated the BDNF receptor TrkB and its downstream signaling, without affecting its other receptor p75NTR. It promoted neurite outgrowth, enhanced spine growth and prevented Aβ-induced cell death in cultured neurons, and facilitated Long-Term Potentiation (LTP) in hippocampal slices. A single-dose tail-vein injection of AS86 activated TrkB signaling in the brain, with a half-life of 6 days in the blood and brain. Bi-weekly peripheral administration of AS86 rescued the deficits in object-recognition memory in the APP/PS1 mouse model. AS86 also reversed spatial memory deficits in the 11-month, but not 14-month old AD mouse model. Conclusion: These results demonstrate the potential of AS86 in AD therapy, suggesting that neuronal and/or synaptic repair as an alternative therapeutic strategy for AD.
Collapse
Affiliation(s)
- Shudan Wang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Hongyang Yao
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
| | - Yihua Xu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Rui Hao
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China, 200065
| | - Wen Zhang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
| | - Hang Liu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Ying Huang
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China, 200065
| | - Wei Guo
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| |
Collapse
|
45
|
The BDNF Val66Met Polymorphism Modulates Resilience of Neurological Functioning to Brain Ageing and Dementia: A Narrative Review. Brain Sci 2020; 10:brainsci10040195. [PMID: 32218234 PMCID: PMC7226504 DOI: 10.3390/brainsci10040195] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023] Open
Abstract
Brain-derived neurotropic factor (BDNF) is an abundant and multi-function neurotrophin in the brain. It is released following neuronal activity and is believed to be particularly important in strengthening neural networks. A common variation in the BDNF gene, a valine to methionine substitution at codon 66 (Val66Met), has been linked to differential expression of BDNF associated with experience-dependent plasticity. The Met allele has been associated with reduced production of BDNF following neuronal stimulation, which suggests a potential role of this variation with respect to how the nervous system may respond to challenges, such as brain ageing and related neurodegenerative conditions (e.g., dementia and Alzheimer’s disease). The current review examines the potential of the BDNF Val66Met variation to modulate an individual’s susceptibility and trajectory through cognitive changes associated with ageing and dementia. On balance, research to date indicates that the BDNF Met allele at this codon is potentially associated with a detrimental influence on the level of cognitive functioning in older adults and may also impart increased risk of progression to dementia. Furthermore, recent studies also show that this genetic variation may modulate an individual’s response to interventions targeted at building cognitive resilience to conditions that cause dementia.
Collapse
|
46
|
Marrocco J, Einhorn NR, Petty GH, Li H, Dubey N, Hoffman J, Berman KF, Goldman D, Lee FS, Schmidt PJ, McEwen BS. Epigenetic intersection of BDNF Val66Met genotype with premenstrual dysphoric disorder transcriptome in a cross-species model of estradiol add-back. Mol Psychiatry 2020; 25:572-583. [PMID: 30356121 PMCID: PMC7042769 DOI: 10.1038/s41380-018-0274-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023]
Abstract
Premenstrual dysphoric disorder (PMDD) affects over 5% of women, with symptoms similar to anxiety and major depression, and is associated with differential sensitivity to circulating ovarian hormones. Little is known about the genetic and epigenetic factors that increase the risk to develop PMDD. We report that 17β-estradiol (E2) affects the behavior and the epigenome in a mouse model carrying a single-nucleotide polymorphism of the brain-derived neurotrophic factor gene (BDNF Val66Met), in a way that recapitulates the hallmarks of PMDD. Ovariectomized mice heterozygous for the BDNF Met allele (Het-Met) and their matched wild-type (WT) mice were administered estradiol or vehicle in drinking water for 6 weeks. Using the open field and the splash test, we show that E2 add-back induces anxiety-like and depression-like behavior in Het-Met mice, but not in WT mice. RNA-seq of the ventral hippocampus (vHpc) highlights that E2-dependent gene expression is markedly different between WT mice and Het-Met mice. Through a comparative whole-genome RNA-seq analysis between mouse vHpc and lymphoblastoid cell line cultures from control women and women with PMDD, we discovered common epigenetic biomarkers that transcend species and cell types. Those genes include epigenetic modifiers of the ESC/E(Z) complex, an effector of response to ovarian steroids. Although the BDNF Met genotype intersects the behavioral and transcriptional traits of women with PMDD, we suggest that these similarities speak to the epigenetic factors by which ovarian steroids produce negative behavioral effects.
Collapse
Affiliation(s)
- Jordan Marrocco
- 0000 0001 2166 1519grid.134907.8Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Nathan R. Einhorn
- 0000 0001 2166 1519grid.134907.8Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Gordon H. Petty
- 0000 0001 2166 1519grid.134907.8Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Howard Li
- 0000 0004 0464 0574grid.416868.5Behavioral Endocrinology Branch, National Institute of Mental Health, Bethesda, MD USA
| | - Neelima Dubey
- grid.440681.fDr. D. Y. Patil Biotechnology & Bioinformatics Institute, Pune, India
| | - Jessica Hoffman
- 0000 0001 0421 5525grid.265436.0Uniformed Services University of the Health Sciences, Bethesda, MD USA
| | - Karen F. Berman
- 0000 0004 0464 0574grid.416868.5Section on Integrative Neuroimaging, National Institute of Mental Health, Bethesda, MD USA
| | - David Goldman
- 0000 0004 0481 4802grid.420085.bLaboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD USA
| | - Francis S. Lee
- 000000041936877Xgrid.5386.8Department of Psychiatry, Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, NY USA
| | - Peter J. Schmidt
- 0000 0004 0464 0574grid.416868.5Behavioral Endocrinology Branch, National Institute of Mental Health, Bethesda, MD USA
| | - Bruce S. McEwen
- 0000 0001 2166 1519grid.134907.8Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| |
Collapse
|
47
|
Laczó J, Cechova K, Parizkova M, Lerch O, Andel R, Matoska V, Kaplan V, Matuskova V, Nedelska Z, Vyhnalek M, Hort J. The Combined Effect of APOE and BDNF Val66Met Polymorphisms on Spatial Navigation in Older Adults. J Alzheimers Dis 2020; 78:1473-1492. [PMID: 33325388 PMCID: PMC7836052 DOI: 10.3233/jad-200615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The apolipoprotein E (APOE) ɛ4 allele is associated with episodic memory and spatial navigation deficits. The brain-derived neurotrophic factor (BDNF) Met allele may further worsen memory impairment in APOEɛ4 carriers but its role in APOEɛ4-related spatial navigation deficits has not been established. OBJECTIVE We examined influence of APOE and BDNF Val66Met polymorphism combination on spatial navigation and volumes of selected navigation-related brain regions in cognitively unimpaired (CU) older adults and those with amnestic mild cognitive impairment (aMCI). METHODS 187 participants (aMCI [n = 116] and CU [n = 71]) from the Czech Brain Aging Study were stratified based on APOE and BDNF Val66Met polymorphisms into four groups: ɛ4-/BDNFVal/Val, ɛ4-/BDNFMet, ɛ4+/BDNFVal/Val, and ɛ4+/BDNFMet. The participants underwent comprehensive neuropsychological examination, brain MRI, and spatial navigation testing of egocentric, allocentric, and allocentric delayed navigation in a real-space human analogue of the Morris water maze. RESULTS Among the aMCI participants, the ɛ4+/BDNFMet group had the least accurate egocentric navigation performance (p < 0.05) and lower verbal memory performance than the ɛ4-/BDNFVal/Val group (p = 0.007). The ɛ4+/BDNFMet group had smaller hippocampal and entorhinal cortical volumes than the ɛ4-/BDNFVal/Val (p≤0.019) and ɛ4-/BDNFMet (p≤0.020) groups. Among the CU participants, the ɛ4+/BDNFMet group had less accurate allocentric and allocentric delayed navigation performance than the ɛ4-/BDNFVal/Val group (p < 0.05). CONCLUSION The combination of APOEɛ4 and BDNF Met polymorphisms is associated with more pronounced egocentric navigation impairment and atrophy of the medial temporal lobe regions in individuals with aMCI and less accurate allocentric navigation in CU older adults.
Collapse
Affiliation(s)
- Jan Laczó
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| | - Katerina Cechova
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| | - Martina Parizkova
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| | - Ondrej Lerch
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| | - Ross Andel
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
- School of Aging Studies, University of South Florida, Tampa, FL, USA
| | - Vaclav Matoska
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Vojtech Kaplan
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Veronika Matuskova
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| | - Zuzana Nedelska
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| | - Martin Vyhnalek
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
48
|
Stonnington CM, Velgos SN, Chen Y, Syed S, Huentelman M, Thiyyagura P, Lee W, Richholt R, Caselli RJ, Locke DE, Lu B, Reiman EM, Su Y, Chen K. Interaction Between BDNF Val66Met and APOE4 on Biomarkers of Alzheimer's Disease and Cognitive Decline. J Alzheimers Dis 2020; 78:721-734. [PMID: 33044176 PMCID: PMC10416650 DOI: 10.3233/jad-200132] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Whether brain-derived neurotrophic factor (BDNF) Met carriage impacts the risk or progression of Alzheimer's disease (AD) is unknown. OBJECTIVE To evaluate the interaction of BDNF Met and APOE4 carriage on cerebral metabolic rate for glucose (CMRgl), amyloid burden, hippocampus volume, and cognitive decline among cognitively unimpaired (CU) adults enrolled in the Arizona APOE cohort study. METHODS 114 CU adults (mean age 56.85 years, 38% male) with longitudinal FDG PET, magnetic resonance imaging, and cognitive measures were BDNF and APOE genotyped. A subgroup of 58 individuals also had Pittsburgh B (PiB) PET imaging. We examined baseline CMRgl, PiB PET amyloid burden, CMRgl, and hippocampus volume change over time, and rate of change in cognition over an average of 15 years. RESULTS Among APOE4 carriers, BDNF Met carriers had significantly increased amyloid deposition and accelerated CMRgl decline in regions typically affected by AD, but without accompanying acceleration of cognitive decline or hippocampal volume changes and with higher baseline frontal CMRgl and slower frontal decline relative to the Val/Val group. The BDNF effects were not found among APOE4 non-carriers. CONCLUSION Our preliminary studies suggest that there is a weak interaction between BDNF Met and APOE4 on amyloid-β plaque burden and longitudinal PET measurements of AD-related CMRgl decline in cognitively unimpaired late-middle-aged and older adults, but with no apparent effect upon rate of cognitive decline. We suggest that cognitive effects of BDNF variants may be mitigated by compensatory increases in frontal brain activity-findings that would need to be confirmed in larger studies.
Collapse
Affiliation(s)
- Cynthia M. Stonnington
- Department of Psychiatry and Psychology, Mayo Clinic Arizona. 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA
| | - Stefanie N. Velgos
- Mayo Clinic Graduate School of Biomedical Sciences, Clinical and Translational Science Track. 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA
- Translational neuroscience and Aging Laboratory, Mayo Clinic Arizona. 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA
| | - Yinghua Chen
- Banner Alzheimer’s Institute. 901 E. Willetta St. Fl 3, Phoenix, AZ 85006, USA
| | - Sameena Syed
- Department of Psychiatry and Psychology, Mayo Clinic Arizona. 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA
- Midwestern University. 19555 N. 59 Ave, Glendale, AZ 85308, USA
- Department of Medicine. University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, OH 44106
| | - Matt Huentelman
- The Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ 85004
| | - Pradeep Thiyyagura
- Banner Alzheimer’s Institute. 901 E. Willetta St. Fl 3, Phoenix, AZ 85006, USA
| | - Wendy Lee
- Banner Alzheimer’s Institute. 901 E. Willetta St. Fl 3, Phoenix, AZ 85006, USA
| | - Ryan Richholt
- The Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ 85004
| | - Richard J. Caselli
- Department of Neurology, Mayo Clinic Arizona. 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA
| | - Dona E.C. Locke
- Department of Psychiatry and Psychology, Mayo Clinic Arizona. 13400 E. Shea Boulevard, Scottsdale, AZ, 85259, USA
| | - Bai Lu
- School of Pharmaceutical Sciences, Tsinghua University. 30 Shuangqing Rd., Haidian Qu, Beijing Shi, China
| | - Eric M. Reiman
- Banner Alzheimer’s Institute. 901 E. Willetta St. Fl 3, Phoenix, AZ 85006, USA
- The Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ 85004
| | - Yi Su
- Banner Alzheimer’s Institute. 901 E. Willetta St. Fl 3, Phoenix, AZ 85006, USA
| | - Kewei Chen
- Banner Alzheimer’s Institute. 901 E. Willetta St. Fl 3, Phoenix, AZ 85006, USA
| |
Collapse
|
49
|
Hardy J, Escott-Price V. Genes, pathways and risk prediction in Alzheimer's disease. Hum Mol Genet 2019; 28:R235-R240. [PMID: 31332445 DOI: 10.1093/hmg/ddz163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/18/2019] [Accepted: 07/04/2019] [Indexed: 11/12/2022] Open
Abstract
The failure of recent clinical trials in Alzheimer's disease has highlighted the need for the development of a more complete understanding of the pathogenesis of the disorder and also a belief that therapies may only work if given very early in the disease process before overt symptoms occur. The rare, early onset forms of the disease are all caused by mutations which make amyloid deposition a more likely event. Here we discuss the recent data showing that, in contrast, much of the risk of late onset disease is encoded by loci involved in lipid metabolism and/or encoded by microglia. We discuss these finding and suggest that amyloid induced membrane damage may be a key factor in disease and also review the evidence that genome wide genetic analysis can substantially help in the prediction of those individuals at high risk of disease in the general population.
Collapse
Affiliation(s)
- John Hardy
- Reta Lilla Research Laboratories, Department of Molecular Neuroscience, University College London Institute of Neurology and UK Dementia Research Institute, University College London, London, UK
| | - Valentina Escott-Price
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences and Dementia Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
50
|
Validation of a priori candidate Alzheimer's disease SNPs with brain amyloid-beta deposition. Sci Rep 2019; 9:17069. [PMID: 31745181 PMCID: PMC6863876 DOI: 10.1038/s41598-019-53604-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
The accumulation of brain amyloid β (Aβ) is one of the main pathological hallmarks of Alzheimer’s disease (AD). However, the role of brain amyloid deposition in the development of AD and the genetic variants associated with this process remain unclear. In this study, we sought to identify associations between Aβ deposition and an a priori evidence based set of 1610 genetic markers, genotyped from 505 unrelated individuals (258 Aβ+ and 247 Aβ−) enrolled in the Australian Imaging, Biomarker & Lifestyle (AIBL) study. We found statistically significant associations for 6 markers located within intronic regions of 6 genes, including AC103796.1-BDNF, PPP3R1, NGFR, KL, ABCA7 & CALHM1. Although functional studies are required to elucidate the role of these genes in the accumulation of Aβ and their potential implication in AD pathophysiology, our findings are consistent with results obtained in previous GWAS efforts.
Collapse
|