1
|
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
|
2
|
Ezzat K, Sturchio A, Espay AJ. The shift to a proteinopenia paradigm in neurodegeneration. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:23-32. [PMID: 36803814 DOI: 10.1016/b978-0-323-85555-6.00001-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
The toxic proteinopathy paradigm has defined neurodegenerative disorders for over a century. This gain-of-function (GOF) framework posited that proteins become toxic when turned into amyloids (pathology), predicting that lowering its levels would translate into clinical benefits. Genetic observations used to support a GOF framework are equally compatible with a loss-of-function (LOF) framework, as the soluble pool of proteins rendered unstable by these mutations (e.g., APP in Alzheimer's disease, SNCA in Parkinson's disease) aggregate, becoming depleted. In this review, we highlight misconceptions that have prevented LOF from gaining currency. Some of these misconceptions include no phenotype in knock-out animals (there is neurodegenerative phenotype in knock-out animals) and high levels of proteins in patients (patients have lower levels of the proteins involved in neurodegeneration than healthy age-matched controls). We also expose the internal contradictions within the GOF framework, namely that (1) pathology can have both pathogenic and protective roles; (2) the neuropathology gold standard for diagnosis can be present in normal individuals and absent in those affected; (3) oligomers are the toxic species even if they are ephemeral and decrease over time. We therefore advocate for a paradigm shift from proteinopathy (GOF) to proteinopenia (LOF) based on the universal depletion of soluble functional proteins in neurodegenerative diseases (low amyloid-β 42 in Alzheimer's disease, low α-synuclein in Parkinson's disease, and low tau in progressive supranuclear palsy) and supported by the confluence of biologic, thermodynamic, and evolutionary principles with proteins having evolved to perform a function, not to become toxic, and where protein depletion is consequential. Such shift to a Proteinopenia paradigm is necessary to examining the safety and efficacy of protein replacement strategies instead of perpetuating a therapeutic paradigm with further antiprotein permutations.
Collapse
Affiliation(s)
- Kariem Ezzat
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Andrea Sturchio
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, Stockholm, Sweden; James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
3
|
Hastings N, Kuan WL, Osborne A, Kotter MRN. Therapeutic Potential of Astrocyte Transplantation. Cell Transplant 2022; 31:9636897221105499. [PMID: 35770772 PMCID: PMC9251977 DOI: 10.1177/09636897221105499] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell transplantation is an attractive treatment strategy for a variety of brain disorders, as it promises to replenish lost functions and rejuvenate the brain. In particular, transplantation of astrocytes has come into light recently as a therapy for amyotrophic lateral sclerosis (ALS); moreover, grafting of astrocytes also showed positive results in models of other conditions ranging from neurodegenerative diseases of older age to traumatic injury and stroke. Despite clear differences in etiology, disorders such as ALS, Parkinson's, Alzheimer's, and Huntington's diseases, as well as traumatic injury and stroke, converge on a number of underlying astrocytic abnormalities, which include inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. In this review, we examine these convergent pathways leading to astrocyte dysfunction, and explore the existing evidence for a therapeutic potential of transplantation of healthy astrocytes in various models. Existing literature presents a wide variety of methods to generate astrocytes, or relevant precursor cells, for subsequent transplantation, while described outcomes of this type of treatment also differ between studies. We take technical differences between methodologies into account to understand the variability of therapeutic benefits, or lack thereof, at a deeper level. We conclude by discussing some key requirements of an astrocyte graft that would be most suitable for clinical applications.
Collapse
Affiliation(s)
- Nataly Hastings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Wei-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Andrew Osborne
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mark R N Kotter
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
4
|
Li D, Zhang J, Li X, Chen Y, Yu F, Liu Q. Insights into lncRNAs in Alzheimer's disease mechanisms. RNA Biol 2021; 18:1037-1047. [PMID: 32605500 PMCID: PMC8216181 DOI: 10.1080/15476286.2020.1788848] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common dementia among the elderly. The pathophysiology of AD is characterized by two hallmarks: amyloid plaques, produced by amyloid β (Aβ) aggregation, and neurofibrillary tangle (NFT), produced by accumulation of phosphorylated tau. The regulatory roles of non-coding RNAs (ncRNAs), particularly long noncoding RNAs (lncRNAs), have been widely recognized in gene expression at the transcriptional and posttranscriptional levels. Mounting evidence shows that lncRNAs are aberrantly expressed in AD progression. Here, we review the lncRNAs that implicated in the regulation of Aβ peptide, tau, inflammation, cell death, and other aspects which are the main mechanisms of AD pathology. We also discuss the possible clinical or therapeutic utility of lncRNA detection or targeting to help diagnose or possibly combat AD.
Collapse
Affiliation(s)
- Dingfeng Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
| | - Juan Zhang
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
| | - Xiaohui Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
| | - Yuhua Chen
- Department of Neurology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Feng Yu
- Department of Neurology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Qiang Liu
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
5
|
Belbin O, Morgan K, Medway C, Warden D, Cortina-Borja M, van Duijn CM, Adams HHH, Frank-Garcia A, Brookes K, Sánchez-Juan P, Alvarez V, Heun R, Kölsch H, Coto E, Kehoe PG, Rodriguez-Rodriguez E, Bullido MJ, Ikram MA, Smith AD, Lehmann DJ. The Epistasis Project: A Multi-Cohort Study of the Effects of BDNF, DBH, and SORT1 Epistasis on Alzheimer's Disease Risk. J Alzheimers Dis 2020; 68:1535-1547. [PMID: 30909233 DOI: 10.3233/jad-181116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pre-synaptic secretion of brain-derived neurotrophic factor (BDNF) from noradrenergic neurons may protect the Alzheimer's disease (AD) brain from amyloid pathology. While the BDNF polymorphism (rs6265) is associated with faster cognitive decline and increased hippocampal atrophy, a replicable genetic association of BDNF with AD risk has yet to be demonstrated. This could be due to masking by underlying epistatic interactions between BDNF and other loci that encode proteins involved in moderating BDNF secretion (DBH and Sortilin). We performed a multi-cohort case-control association study of the BDNF, DBH, and SORT1 loci comprising 5,682 controls and 2,454 AD patients from Northern Europe (87% of samples) and Spain (13%). The BDNF locus was associated with increased AD risk (odds ratios; OR = 1.1-1.2, p = 0.005-0.3), an effect size that was consistent in the Northern European (OR = 1.1-1.2, p = 0.002-0.8) but not the smaller Spanish (OR = 0.8-1.6, p = 0.4-1.0) subset. A synergistic interaction between BDNF and sex (synergy factor; SF = 1.3-1.5 p = 0.002-0.02) translated to a greater risk of AD associated with BDNF in women (OR = 1.2-1.3, p = 0.007-0.00008) than men (OR = 0.9-1.0, p = 0.3-0.6). While the DBH polymorphism (rs1611115) was also associated with increased AD risk (OR = 1.1, p = 0.04) the synergistic interaction (SF = 2.2, p = 0.007) between BDNF (rs6265) and DBH (rs1611115) contributed greater AD risk than either gene alone, an effect that was greater in women (SF = 2.4, p = 0.04) than men (SF = 2.0, p = 0.2). These data support a complex genetic interaction at loci encoding proteins implicated in the DBH-BDNF inflammatory pathway that modifies AD risk, particularly in women.
Collapse
Affiliation(s)
- Olivia Belbin
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Kevin Morgan
- Human Genetics School of Life Sciences, University of Nottingham, UK
| | - Chris Medway
- Institute of Medical Genetics, University Hospital Wales, Cardiff, UK
| | - Donald Warden
- Oxford Project to Investigate Memory and Ageing (OPTIMA), University Department of Pharmacology, Oxford, UK
| | - Mario Cortina-Borja
- Clinical Epidemiology, Nutrition and Biostatistics, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Ana Frank-Garcia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain
| | - Keeley Brookes
- Human Genetics School of Life Sciences, University of Nottingham, UK
| | - Pascual Sánchez-Juan
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Victoria Alvarez
- Laboratorio de Genética, AGC Laboratorio de Medicina, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Reinhard Heun
- Department of Psychiatry, University of Bonn, Bonn, Germany
| | - Heike Kölsch
- Department of Psychiatry, University of Bonn, Bonn, Germany
| | - Eliecer Coto
- Laboratorio de Genética, AGC Laboratorio de Medicina, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Patrick G Kehoe
- Dementia Research Group, Bristol Medical School Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol, UK
| | - Eloy Rodriguez-Rodriguez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Maria J Bullido
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - A David Smith
- Oxford Project to Investigate Memory and Ageing (OPTIMA), University Department of Pharmacology, Oxford, UK
| | - Donald J Lehmann
- Oxford Project to Investigate Memory and Ageing (OPTIMA), University Department of Pharmacology, Oxford, UK
| |
Collapse
|
6
|
Balietti M, Giuli C, Casoli T, Fabbietti P, Conti F. Is Blood Brain-Derived Neurotrophic Factor a Useful Biomarker to Monitor Mild Cognitive Impairment Patients? Rejuvenation Res 2020; 23:411-419. [PMID: 32200710 DOI: 10.1089/rej.2020.2307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Availability of reliable prognostic biomarkers that are also able to monitor preventive/therapeutic interventions in patients with mild cognitive impairment (MCI) is crucial. Cerebral brain-derived neurotrophic factor (BDNF) alterations were evidenced in Alzheimer's disease, but the value of blood BDNF in MCI is unclear, especially because of the incomplete/incorrect management of the numerous confounding factors unrelated to the disease. This study, applying a multidisciplinary methodological approach, aimed at clarifying whether blood BDNF can really mirror the cognitive symptoms of MCI, thus supporting the evaluation of clinical protocols' effectiveness as well as the definition of the conversion rate to dementia. Healthy elderly subjects (HE) and MCI patients were assessed for sociodemographic, neuropsychological, pharmacological, and lifestyle data, and plasma BDNF was measured (baseline); then, in the MCI cohort, the biomarker was tested in a comprehensive cognitive stimulation intervention (CS) as well as in a 2-year follow-up period. Plasma BDNF, cleansed from all the interfering factors, (1) did not discriminate HE and MCI patients; (2) in MCI patients reflected mood, social engagement, and subjective memory complaints but not cognition; (3) changed due to CS, although with no correlations to cognitive performances; and (4) predicted no functional deterioration. Our data indicate that the possible biased use of plasma BDNF in MCI is critically risky.
Collapse
Affiliation(s)
- Marta Balietti
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Cinzia Giuli
- Geriatrics Operative Unit, IRCCS INRCA, Fermo, Italy
| | - Tiziana Casoli
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Paolo Fabbietti
- Unit of Geriatric Pharmacoepidemiology, IRCCS INRCA, Ancona, Italy
| | - Fiorenzo Conti
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.,Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
7
|
Chen X, Li Z, Cheng Y, Kardami E, Loh YP. Low and High Molecular Weight FGF-2 Have Differential Effects on Astrocyte Proliferation, but Are Both Protective Against Aβ-Induced Cytotoxicity. Front Mol Neurosci 2020; 12:328. [PMID: 32038161 PMCID: PMC6992557 DOI: 10.3389/fnmol.2019.00328] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 12/23/2019] [Indexed: 12/17/2022] Open
Abstract
Astrocytes are the most abundant type of glial cells in the brain, and they play a key role in Alzheimer’s disease (AD). Fibroblast Growth Factor-2 (FGF-2) has been implicated as a potential therapeutic agent for treating AD. In the present study, we investigated the protective effects of low molecular weight (LMW; 17 KDa) and high molecular weight (HMW; 23 KDa) forms of FGF-2 on Aβ1–42-induced toxicity, and proliferation in astrocytes. We show that both isoforms of FGF-2 have similar protective effects against Aβ1–42-induced cytotoxicity in primary cultured cortical astrocytes as measured by Lactate Dehydrogenase (LDH) release assay. Additionally, 17 KDa FGF-2 significantly promoted astrocyte proliferation as measured by Trypan Blue, DRAQ5 and 5-ethynyl-2’-deoxyuridine (EdU) staining, but not 23 kDa FGF-2. Furthermore, our results demonstrated that AKT signaling pathway was required for the protective and proliferative effects of FGF-2. Downstream effector studies indicated that 17 kDa FGF-2 promoted astrocyte proliferation by enhanced expression of c-Myc, Cyclin D1, Cyclin E. Furthermore, our data suggested that Cyclin D1 was required for the proliferative effect of LMW FGF2 in astrocytes. Taken together, our findings provide important information for the similarities and differences between 23 kDa and17 kDa isoforms of FGF-2 on astrocyte survival and proliferation.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Zhaojin Li
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Yong Cheng
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China.,Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Elissavet Kardami
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Y Peng Loh
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
8
|
Enette L, Vogel T, Merle S, Valard-Guiguet AG, Ozier-Lafontaine N, Neviere R, Leuly-Joncart C, Fanon JL, Lang PO. Effect of 9 weeks continuous vs. interval aerobic training on plasma BDNF levels, aerobic fitness, cognitive capacity and quality of life among seniors with mild to moderate Alzheimer's disease: a randomized controlled trial. Eur Rev Aging Phys Act 2020; 17:2. [PMID: 31921371 PMCID: PMC6945614 DOI: 10.1186/s11556-019-0234-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 12/29/2019] [Indexed: 12/19/2022] Open
Abstract
Background Evidence suggests that aerobic-type training confers physical benefits and appears to contribute positively to brain health. This study aims to compare the effect of 9-weeks continuous (CAT) to interval aerobic training (IAT) on brain derived neurotrophic factor (BDNF) plasma level, aerobic fitness, cognitive performance, and quality of life among senior with Alzheimer's disease (AD). Methods 52 participants were randomly allocated into three groups (CAT n = 14; IAT n = 17; and Controls n = 21). CAT and IAT consisted of 18 sessions of 30-min cycling, twice a week, over 9 weeks. During the same period, controls were engaged in interactive information sessions. Plasma BDNF level; aerobic fitness parameters (Metabolic equivalent task - METs; Maximal Tolerated Power - MTP); functional capacities (6-Minute Walk Test - 6MWT); cognitive performance (Mini Mental State Examination; Rey auditory verbal learning test; and digit span test) and quality of life (Quality Of Life of Alzheimer's Disease scale - QoL-AD) were measured in all participants at baseline and 9 weeks later. A third plasma BDNF level was quantified following a 4 weeks detraining. Results No significant change was measured in terms of plasma BDNF level and cognitive performance after interventions, in all groups compared to baseline. After 9 weeks, CAT and IAT significantly improved aerobic fitness parameters compared to controls (METs: + 0.6 and + 1.0 vs. + 0.4; MTP: + 16 watts and + 20 watts vs. + 10 watts; and functional capacities (6MWT: + 22 m and + 31 m vs. -40 m). Compared to controls, QoL-AD after CAT was improved (+ 2 points; p = 0.02). Conclusions Neither aerobic exercise modalities significantly modified plasma BDNF levels and cognitive performances. CAT and IAT enhanced aerobic fitness and functional capacities in AD patients and CAT their QoL. Trial registration ClinicalTrials.gov website (NCT02968875); registration date: 7 September 2016. "Retrospectively registered".
Collapse
Affiliation(s)
- Lievyn Enette
- 1Research Laboratory Mitochondria, Oxidative stress and muscle resistance (MSP, EA-3072), Department of Physiology, Faculty of Medicine, Strasbourg University, Résidence La Yole, bat. B L'Etang Z'abricot, 97200 Strasbourg, France
| | - Thomas Vogel
- 1Research Laboratory Mitochondria, Oxidative stress and muscle resistance (MSP, EA-3072), Department of Physiology, Faculty of Medicine, Strasbourg University, Résidence La Yole, bat. B L'Etang Z'abricot, 97200 Strasbourg, France.,2Department of geriatric, University Hospital, Strasbourg, France
| | - Sylvie Merle
- Methodology and biostatistics Unit (DRCI), University Hospital Centre of Martinique, Fort de France, France
| | - Anna-Gaelle Valard-Guiguet
- The Caribbean reference center for rare neuromuscular and neurologic diseases (CeRCa), University Hospital Centre of Martinique, Fort de France, France
| | - Nathalie Ozier-Lafontaine
- Department of Functional Exploration and Non-Invasive Cardiology, University Hospital Centre of Martinique, Fort de France, France
| | - Remi Neviere
- Department of cardiology, University Hospital Centre of Martinique, Fort de France, France
| | - Claudia Leuly-Joncart
- Department of Geriatric and Gerontology, University Hospital Centre of Martinique, Fort de France, France
| | - Jean Luc Fanon
- Department of Geriatric and Gerontology, University Hospital Centre of Martinique, Fort de France, France
| | | |
Collapse
|
9
|
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: 16] [Impact Index Per Article: 3.2] [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
|
10
|
Noris-García E, Arce S, Nardin P, Lanigan ME, Acuña V, Gutierrez F, Robinson-Agramonte MA, Gonçalves CA. Peripheral levels of brain-derived neurotrophic factor and S100B in neuropsychiatric systemic lupus erythematous. Lupus 2018; 27:2041-2049. [PMID: 30376438 DOI: 10.1177/0961203318804899] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The aim of this study was to investigate serum S100B and brain-derived neurotrophic factor (BDNF) in systemic lupus erythematous (SLE) patients, with and without neuropsychiatric (NP) manifestation activity. METHODS We assessed 47 SLE patients and 20 selected healthy individuals. Disease activity was assessed according to the SLE disease activity index (SLEDAI). Serum BDNF and S100B were measured by enzyme-linked immunosorbent assay. RESULTS Serum S100B protein was significantly higher in SLE patients. BDNF levels were significantly decreased in active SLE, when compared with inactive SLE, but not when compared with controls. S100B was clearly higher in the NPSLE group, when compared with the non-NPSLE or control groups. Receiver operating characteristic analysis of S100B revealed an area under the curve of 0.706 that discriminated NPSLE patients with peripheral polyneuropathy. CONCLUSIONS Our findings reinforce the use of serum S100B as a biomarker in SLE, particularly for NPSLE. Moreover, we found a strong association between serum S100B and peripheral neuropathy, indicating a specific utility for this biomarker in SLE that warrants clinical investigation.
Collapse
Affiliation(s)
- E Noris-García
- 1 Department of Immunology, Nephrology Institute, Havana, Cuba
| | - S Arce
- 1 Department of Immunology, Nephrology Institute, Havana, Cuba
| | - P Nardin
- 2 Faculty of Pharmaceutical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - M E Lanigan
- 3 Department of Psychiatric, Joaquín Albarrán Hospital, Havana, Cuba
| | - V Acuña
- 3 Department of Psychiatric, Joaquín Albarrán Hospital, Havana, Cuba
| | - F Gutierrez
- 3 Department of Psychiatric, Joaquín Albarrán Hospital, Havana, Cuba
| | - M A Robinson-Agramonte
- 4 Department of Neuroimmunology, International Center for Neurological Restoration, Havana, Cuba
| | - C-A Gonçalves
- 5 Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
11
|
Lim YY, Hassenstab J, Goate A, Fagan AM, Benzinger TLS, Cruchaga C, McDade E, Chhatwal J, Levin J, Farlow MR, Graff-Radford NR, Laske C, Masters CL, Salloway S, Schofield P, Morris JC, Maruff P, Bateman RJ. Effect of BDNFVal66Met on disease markers in dominantly inherited Alzheimer's disease. Ann Neurol 2018; 84:424-435. [PMID: 30014553 DOI: 10.1002/ana.25299] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Previous studies suggest that the brain-derived neurotrophic factor (BDNF) Val66Met (rs6265) polymorphism may influence symptom onset in Alzheimer's disease (AD). Our recent cross-sectional findings suggest that Met66 may influence clinical expression in dominantly inherited AD (DIAD) through its effects on tau. However, it remains unclear whether carriage of Met66 in DIAD results in faster increases in cerebrospinal fluid (CSF) tau and ptau181 , and whether these increases are associated with accelerated brain volume loss and memory decline. METHODS A total of 211 subjects (101 mutation noncarriers, 110 mutation carriers), who were cognitively normal, as defined by a Clinical Dementia Rating global score of 0, completed assessments of cognitive function, neuroimaging, and CSF sampling over 3.5 years as part of the Dominantly Inherited Alzheimer's Network. RESULTS In mutation carriers, Met66 carriers showed faster memory decline (4×), hippocampal volume loss (16×), and CSF tau and ptau181 increases (6×) than Val66 homozygotes. BDNF did not influence rates of cortical β-amyloid accumulation or change in CSF Aβ42 levels in mutation carriers. In mutation noncarriers, BDNF genotype had no effect on change in cognition, brain volume, cortical β-amyloid accumulation, or change in any CSF measures of tau, ptau181 , and CSF Aβ42 . INTERPRETATION As in sporadic AD, the deleterious effects of β-amyloid on cognitive function, brain volume loss, and CSF tau in DIAD mutation carriers are less in Val66 homozygotes. The BDNF Val66Met polymorphism should be considered as a potential moderator of clinical trial outcomes in current treatment and prevention trials in DIAD and sporadic AD. Ann Neurol 2018;84:424-435.
Collapse
Affiliation(s)
- Yen Ying Lim
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Jason Hassenstab
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anne M Fagan
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | | | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Johannes Levin
- Department of Neurology, University of Munich, Munich, Germany
| | - Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN
| | | | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Section for Dementia Research, Department of Cellular Neurology, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Stephen Salloway
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI
| | - Peter Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Cogstate Ltd, Melbourne, Victoria, Australia
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | | |
Collapse
|
12
|
Balietti M, Giuli C, Conti F. Peripheral Blood Brain-Derived Neurotrophic Factor as a Biomarker of Alzheimer's Disease: Are There Methodological Biases? Mol Neurobiol 2018; 55:6661-6672. [PMID: 29330839 PMCID: PMC6061178 DOI: 10.1007/s12035-017-0866-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Abstract
Mounting evidence that alterations in brain-derived neurotrophic factor (BDNF) levels and signaling may be involved in the etiopathogenesis of Alzheimer's disease (AD) has suggested that its blood levels could be used as a biomarker of the disease. However, higher, lower, or unchanged circulating BDNF levels have all been described in AD patients compared to healthy controls. Although the reasons for such different findings are unclear, methodological issues are likely to be involved. The heterogeneity of participant recruitment criteria and the lack of control of variables that influence circulating BDNF levels regardless of dementia (depressive symptoms, medications, lifestyle, lack of overlap between serum and plasma, and experimental aspects) are likely to bias result and prevent study comparability. The present work reviews a broad panel of factors, whose close control could help reduce the inconsistency of study findings, and offers practical advice on their management. Research directed at elucidating the weight of each of these variables and at standardizing analytical methodologies is urgently needed.
Collapse
Affiliation(s)
- Marta Balietti
- Center for Neurobiology of Aging, INRCA, Via Birarelli 8, 60121, Ancona, Italy.
| | - Cinzia Giuli
- Geriatrics Operative Unit, INRCA, Fermo, 63023, Italy
| | - Fiorenzo Conti
- Center for Neurobiology of Aging, INRCA, Via Birarelli 8, 60121, Ancona, Italy
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, 60126, Italy
| |
Collapse
|
13
|
Merlo S, Spampinato SF, Beneventano M, Sortino MA. The contribution of microglia to early synaptic compensatory responses that precede β-amyloid-induced neuronal death. Sci Rep 2018; 8:7297. [PMID: 29740062 PMCID: PMC5940848 DOI: 10.1038/s41598-018-25453-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/17/2018] [Indexed: 12/31/2022] Open
Abstract
Glial-neuronal cross-talk has a critical role in the development of neurodegenerative conditions, including Alzheimer's Disease, where it affects neuronal responses to β-amyloid peptide (Aβ)-induced toxicity. We set out to identify factors regulating synaptic responses to Aβ, dissecting the specific role of glial signaling. A low concentration of aggregated Aβ42 induced selective up-regulation of mature brain-derived neurotrophic factor (BDNF) expression and release in rat organotypic hippocampal cultures as well as in cortical pure microglia. Conditioned media from resting (CMC) or Aβ42-treated (CMA) microglia were tested for their effects on synaptophysin expression in SH-SY5Y neuronal-like cells during challenge with Aβ42. Both CMC and CMA prevented Aβ-induced synaptophysin loss. In the presence of Aβ + CMA, synaptophysin was over-expressed, although it appeared partly clumped in cell bodies. Synaptophysin over-expression was not directly dependent on BDNF signaling on neuronal-like cells, but relied on autocrine BDNF action on microglia. FM1-43 labeling experiments revealed compromised synaptic vesicle recycling in Aβ42-treated neuronal-like cells, rescued by microglial conditioned medium. In these conditions, significant and prolonged neuroprotection was observed. Our results point to microglia as a target for early intervention, given its positive role in supporting neuronal compensatory responses to Aβ synaptotoxicity, which potentially lead to their extended survival.
Collapse
Affiliation(s)
- Sara Merlo
- Department of Biomedical and Biotechnological Sciences, section of Pharmacology, University of Catania, Catania, Italy
| | - Simona Federica Spampinato
- Department of Biomedical and Biotechnological Sciences, section of Pharmacology, University of Catania, Catania, Italy
| | - Martina Beneventano
- Department of Biomedical and Biotechnological Sciences, section of Pharmacology, University of Catania, Catania, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, section of Pharmacology, University of Catania, Catania, Italy.
| |
Collapse
|
14
|
Balietti M, Giuli C, Fattoretti P, Fabbietti P, Papa R, Postacchini D, Conti F. Effect of a Comprehensive Intervention on Plasma BDNF in Patients with Alzheimer's Disease. J Alzheimers Dis 2018; 57:37-43. [PMID: 28222525 PMCID: PMC5345639 DOI: 10.3233/jad-161168] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A comprehensive intervention (CI) on patients with Alzheimer’s disease was assessed by measuring plasmabrain-derived neurotrophic factor (pBDNF) and ADAS-Cog score (ADAS-Cogscore) before, immediately after (FU1), and 6 (FU2) and 24 months (FU3) after the CI. Baseline pBDNF was higher in patients with moderate AD (but not mild AD) than in healthy controls. At FU1, pBDNF and ADAS-Cogscore decreased significantly. At FU2 and FU3, patients’ cognitive status worsened and pBDNF further increased versus baseline, suggesting that CI interruption may be a stress event that prevents return to homeostasis. CI exerted positive short-term effects, but more information is needed on long-term consequences.
Collapse
Affiliation(s)
| | | | | | | | - Roberta Papa
- Center of Socio-economic Gerontological Research, INRCA, Ancona, Italy
| | | | - Fiorenzo Conti
- Center for Neurobiology of Aging, INRCA, Ancona, Italy.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
15
|
Benussi L, Binetti G, Ghidoni R. Loss of Neuroprotective Factors in Neurodegenerative Dementias: The End or the Starting Point? Front Neurosci 2017; 11:672. [PMID: 29249935 PMCID: PMC5717017 DOI: 10.3389/fnins.2017.00672] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/20/2017] [Indexed: 01/05/2023] Open
Abstract
Recent clinical, genetic and biochemical experimental evidences highlight the existence of common molecular pathways underlying neurodegenerative diseases. In this review, we will explore a key common pathological mechanism, i.e., the loss of neuroprotective factors, across the three major neurodegenerative diseases leading to dementia: Alzheimer's disease (AD), Frontotemporal dementia (FTD) and Lewy body dementia (LBD). We will report evidences that the Brain Derived Neurotrophic Factor (BDNF), the most investigated and characterized brain neurotrophin, progranulin, a multi-functional adipokine with trophic and growth factor properties, and cystatin C, a neuroprotective growth factor, are reduced in AD, FTD, and LBD. Moreover, we will review the molecular mechanism underlying the loss of neuroprotective factors in neurodegenerative diseases leading to dementia, with a special focus on endo-lysosomal pathway and intercellular communication mediated by extracellular vesicles. Exploring the shared commonality of disease mechanisms is of pivotal importance to identify novel potential therapeutic targets and to develop treatments to delay, slow or block disease progression.
Collapse
Affiliation(s)
- Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,MAC Memory Center, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
16
|
Boots EA, Schultz SA, Clark LR, Racine AM, Darst BF, Koscik RL, Carlsson CM, Gallagher CL, Hogan KJ, Bendlin BB, Asthana S, Sager MA, Hermann BP, Christian BT, Dubal DB, Engelman CD, Johnson SC, Okonkwo OC. BDNF Val66Met predicts cognitive decline in the Wisconsin Registry for Alzheimer's Prevention. Neurology 2017; 88:2098-2106. [PMID: 28468845 DOI: 10.1212/wnl.0000000000003980] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 03/13/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To examine the influence of the brain-derived neurotrophic factor (BDNF) Val66Met polymorphism on longitudinal cognitive trajectories in a large, cognitively healthy cohort enriched for Alzheimer disease (AD) risk and to understand whether β-amyloid (Aβ) burden plays a moderating role in this relationship. METHODS One thousand twenty-three adults (baseline age 54.94 ± 6.41 years) enrolled in the Wisconsin Registry for Alzheimer's Prevention underwent BDNF genotyping and cognitive assessment at up to 5 time points (average follow-up 6.92 ± 3.22 years). A subset (n = 140) underwent 11C-Pittsburgh compound B (PiB) scanning. Covariate-adjusted mixed-effects regression models were used to elucidate the effect of BDNF on cognitive trajectories in 4 cognitive domains, including verbal learning and memory, speed and flexibility, working memory, and immediate memory. Secondary mixed-effects regression models were conducted to examine whether Aβ burden, indexed by composite PiB load, modified any observed BDNF-related cognitive trajectories. RESULTS Compared to BDNF Val/Val homozygotes, Met carriers showed steeper decline in verbal learning and memory (p = 0.002) and speed and flexibility (p = 0.017). In addition, Aβ burden moderated the relationship between BDNF and verbal learning and memory such that Met carriers with greater Aβ burden showed even steeper cognitive decline (p = 0.033). CONCLUSIONS In a middle-aged cohort with AD risk, carriage of the BDNF Met allele was associated with steeper decline in episodic memory and executive function. This decline was exacerbated by greater Aβ burden. These results suggest that the BDNF Val66Met polymorphism may play an important role in cognitive decline and could be considered as a target for novel AD therapeutics.
Collapse
Affiliation(s)
- Elizabeth A Boots
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Stephanie A Schultz
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Lindsay R Clark
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Annie M Racine
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Burcu F Darst
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Rebecca L Koscik
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Cynthia M Carlsson
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Catherine L Gallagher
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Kirk J Hogan
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Barbara B Bendlin
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Sanjay Asthana
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Mark A Sager
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Bruce P Hermann
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Bradley T Christian
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Dena B Dubal
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Corinne D Engelman
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Sterling C Johnson
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco
| | - Ozioma C Okonkwo
- From the Geriatric Research Education and Clinical Center (E.A.B., S.A.S., L.R.C., C.M.C., C.L.G., B.B.B., S.A., S.C.J., O.C.C.), William S. Middleton Memorial Veterans Hospital; Wisconsin Alzheimer's Disease Research Center (E.A.B., S.A.S., L.R.C., A.M.R., C.M.C., C.L.G., B.B.B., S.A., M.A.S., B.P.H., B.T.C., S.C.J., O.C.O.), Wisconsin Alzheimer's Institute (L.R.C., R.L.K., C.M.C., K.J.H., B.B.B., S.A., M.A.S., B.P.H., C.D.E., S.C.J., O.C.O.), Department of Population Health Sciences (B.F.D., C.D.E.), Department of Neurology (C.L.G., B.P.H.), Department of Anesthesiology (K.J.H.), Department of Radiology (M.A.S.), and Department of Medical Physics (B.T.C.), University of Wisconsin School of Medicine and Public Health, Madison; and Department of Neurology (D.B.D.), University of California, San Francisco.
| |
Collapse
|
17
|
Janssens J, Lu D, Ni B, Chadwick W, Siddiqui S, Azmi A, Etienne H, Jushaj A, van Gastel J, Martin B, Maudsley S. Development of Precision Small-Molecule Proneurotrophic Therapies for Neurodegenerative Diseases. VITAMINS AND HORMONES 2016; 104:263-311. [PMID: 28215298 DOI: 10.1016/bs.vh.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Age-related neurodegenerative diseases, such as Alzheimer's disease, will represent one of the largest future burdens on worldwide healthcare systems due to the increasing proportion of elderly in our society. As deficiencies in neurotrophins are implicated in the pathogenesis of many age-related neurodegenerative disorders, it is reasonable to consider that global neurotrophin resistance may also become a major healthcare threat. Central nervous system networks are effectively maintained through aging by neuroprotective and neuroplasticity signaling mechanisms which are predominantly controlled by neurotrophin receptor signaling. Neurotrophin receptors are single pass receptor tyrosine kinases that form dimeric structures upon ligand binding to initiate cellular signaling events that control many protective and plasticity-related pathways. Declining functionality of the neurotrophin ligand-receptor system is considered one of the hallmarks of neuropathological aging. Therefore, it is imperative to develop effective therapeutic strategies to contend with this significant issue. While the therapeutic applications of cognate ligands for neurotrophin receptors are limited, the development of nonpeptidergic, small-molecule ligands can overcome these limitations, and productively regulate this important receptor system with beneficial effects. Using our advanced knowledge of the high-dimensionality complexity of receptor systems, the future generation of precision medicines targeting these systems will be an attainable goal.
Collapse
Affiliation(s)
- J Janssens
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - D Lu
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - B Ni
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - W Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Siddiqui
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - A Azmi
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - H Etienne
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - A Jushaj
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - J van Gastel
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - B Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Maudsley
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium; Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States.
| |
Collapse
|
18
|
Central visual pathways in glaucoma: evidence for distal mechanisms of neuronal self-repair. J Neuroophthalmol 2016; 35 Suppl 1:S29-37. [PMID: 26274834 DOI: 10.1097/wno.0000000000000291] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
As in other age-related neurodegenerative diseases, progression of neurodegeneration in glaucoma involves early axonopathy. In glaucoma, this is marked by degradation of active transport along retinal ganglion cell (RGC) axons projecting from the retina to the brain. In experimental systems, transport degradation occurs first in the most distal site in the RGC projection, the superior colliculus (SC) of the midbrain. Even as degradation progresses from one retinotopic sector to the next, important structures in the affected sectors persist, including synapses from RGC axon terminals onto SC neurons. This structural persistence is accompanied by focally increased brain-derived neurotrophic factor in hypertrophic SC astrocyte glia and defines a therapeutic window of opportunity. Thus, central brain structures in glaucoma may respond to disease-relevant stress by induction of mechanisms useful for maintaining retinal signals.
Collapse
|
19
|
Treatment with the neurotoxic Aβ (25-35) peptide modulates the expression of neuroprotective factors Pin1, Sirtuin 1, and brain-derived neurotrophic factor in SH-SY5Y human neuroblastoma cells. ACTA ACUST UNITED AC 2016; 68:271-6. [PMID: 26915812 DOI: 10.1016/j.etp.2016.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/12/2016] [Accepted: 02/15/2016] [Indexed: 12/12/2022]
Abstract
The deposition of Amyloid β peptide plaques is a pathological hallmark of Alzheimer's disease (AD). The Aβ (25-35) peptide is regarded as the toxic fragment of full-length Aβ (1-42). The mechanism of its toxicity is not completely understood, along with its contribution to AD pathological processes. The aim of this study was to investigate the effect of the neurotoxic Aβ (25-35) peptide on the expression of the neuroprotective factors Pin1, Sirtuin1, and Bdnf in human neuroblastoma cells. Levels of Pin1, Sirtuin 1, and Bdnf were compared by real-time PCR and Western blotting in SH-SY5Y cells treated with Aβ (25-35) or administration vehicle. The level of Pin1 gene and protein expression was significantly decreased in cells exposed to 25 μM Aβ (25-35) compared to vehicle-treated controls. Similarly, Sirtuin1 expression was significantly reduced by Aβ (25-35) exposure. In contrast, both Bdnf mRNA and protein levels were significantly increased by Aβ (25-35) treatment, suggesting the activation of a compensatory response to the insult. Both Pin1 and Sirtuin 1 exert a protective role by reducing the probability of plaque deposition, since they promote amyloid precursor protein processing through non-amyloidogenic pathways. The present results show that Aβ (25-35) peptide reduced the production of these neuroprotective proteins, thus further increasing Aβ generation.
Collapse
|
20
|
Xu Z, Xiao N, Chen Y, Huang H, Marshall C, Gao J, Cai Z, Wu T, Hu G, Xiao M. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Aβ accumulation and memory deficits. Mol Neurodegener 2015; 10:58. [PMID: 26526066 PMCID: PMC4631089 DOI: 10.1186/s13024-015-0056-1] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/29/2015] [Indexed: 12/20/2022] Open
Abstract
Background Preventing or reducing amyloid-beta (Aβ) accumulation in the brain is an important therapeutic strategy for Alzheimer’s disease (AD). Recent studies showed that the water channel aquaporin-4 (AQP4) mediates soluble Aβ clearance from the brain parenchyma along the paravascular pathway. However the direct evidence for roles of AQP4 in the pathophysiology of AD remains absent. Results Here, we reported that the deletion of AQP4 exacerbated cognitive deficits of 12-moth old APP/PS1 mice, with increases in Aβ accumulation, cerebral amyloid angiopathy and loss of synaptic protein and brain-derived neurotrophic factor in the hippocampus and cortex. Furthermore, AQP4 deficiency increased atrophy of astrocytes with significant decreases in interleukin-1 beta and nonsignficant decreases in interleukin-6 and tumor necrosis factor-alpha in hippocampal and cerebral samples. Conclusions These results suggest that AQP4 attenuates Aβ pathogenesis despite its potentially inflammatory side-effects, thus serving as a promising target for treating AD. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0056-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhiqiang Xu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu, 210029, China
| | - Na Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu, 210029, China
| | - Yali Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu, 210029, China
| | - Huang Huang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu, 210029, China
| | - Charles Marshall
- Department of Rehabilitation Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, USA
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu, 210029, China
| | - Zhiyou Cai
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ting Wu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu, 210029, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
21
|
Hall VJ, Lindblad MM, Jakobsen JE, Gunnarsson A, Schmidt M, Rasmussen MA, Volke D, Zuchner T, Hyttel P. Impaired APP activity and altered Tau splicing in embryonic stem cell-derived astrocytes obtained from an APPsw transgenic minipig. Dis Model Mech 2015; 8:1265-78. [PMID: 26398935 PMCID: PMC4610230 DOI: 10.1242/dmm.019489] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 07/27/2015] [Indexed: 01/09/2023] Open
Abstract
Animal models of familial juvenile onset of Alzheimer's disease (AD) often fail to produce diverse pathological features of the disease by modification of single gene mutations that are responsible for the disease. They can hence be poor models for testing and development of novel drugs. Here, we analyze in vitro-produced stem cells and their derivatives from a large mammalian model of the disease created by overexpression of a single mutant human gene (APPsw). We produced hemizygous and homozygous radial glial-like cells following culture and differentiation of embryonic stem cells (ESCs) isolated from embryos obtained from mated hemizygous minipigs. These cells were confirmed to co-express varying neural markers, including NES, GFAP and BLBP, typical of type one radial glial cells (RGs) from the subgranular zone. These cells had altered expression of CCND1 and NOTCH1 and decreased expression of several ribosomal RNA genes. We found that these cells were able to differentiate into astrocytes upon directed differentiation. The astrocytes produced had decreased α- and β-secretase activity, increased γ-secretase activity and altered splicing of tau. This indicates novel aspects of early onset mechanisms related to cell renewal and function in familial AD astrocytes. These outcomes also highlight that radial glia could be a potentially useful population of cells for drug discovery, and that altered APP expression and altered tau phosphorylation can be detected in an in vitro model of the disease. Finally, it might be possible to use large mammal models to model familial AD by insertion of only a single mutation. Summary: Insight into astrocyte and radial glia pathology in an in vitro culture system derived from the APPsw pig.
Collapse
Affiliation(s)
- Vanessa J Hall
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | - Maiken M Lindblad
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | - Jannik E Jakobsen
- Department of Biomedicine, Aarhus University, Faculty of Health, DK-8000 Aarhus, Denmark
| | - Anders Gunnarsson
- Department of Biomedicine, Aarhus University, Faculty of Health, DK-8000 Aarhus, Denmark
| | - Mette Schmidt
- Department of Large Animal Sciences, Faculty of Life Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | | | - Daniela Volke
- Center for Biotechnology and Biomedicine, Institute of Bioanalytical Chemistry, University of Leipzig, 04103 Leipzig, Germany
| | - Thole Zuchner
- Octapharma Biopharmaceuticals GmbH, 69120 Heidelberg, Germany
| | - Poul Hyttel
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| |
Collapse
|
22
|
Plasma BDNF levels associate with Pittsburgh compound B binding in the brain. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2015. [PMID: 26207261 PMCID: PMC4507280 DOI: 10.1016/j.dadm.2015.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Brain-derived neurotrophic factor (BDNF) plays an important role in Alzheimer's disease (AD) and other neurodegenerative disorders. BDNF function is adversely affected by amyloid beta in AD. BDNF levels in brain and peripheral tissues are lower in patients with AD and mild cognitive impairment (MCI) than in controls. Here we examined the association between plasma levels of BDNF and amyloid deposition in the brain measured with Pittsburgh Compound B (PiB). Method Our data set consisted of 18 AD, 56 MCI, and 3 normal control Alzheimer's Disease Neuroimaging Initiative-1 (ADNI1) subjects with available [11C] PiB and peripheral blood protein data. Magnetic resonance imaging (MRI)-coregistered positron emission tomography data were smoothed with a 15 mm kernel and mapped onto three-dimensional (3D) hemispheric models using the warping deformations computed in cortical pattern matching of the associated MRI scans. We applied linear regression to examine in 3D the associations between BDNF and PiB standard uptake value ratio, while adjusting for age and sex. We used permutation statistics thresholded at P < .01 for multiple comparisons correction. Results Plasma BDNF levels showed significant negative associations with left greater than right amyloid burden in the lateral temporal, inferior parietal, inferior frontal, anterior and posterior cingulate, and orbitofrontal regions (left Pcorrected = .03). Conclusions As hypothesized, lower plasma levels of BDNF were significantly associated with widespread brain amyloidosis.
Collapse
|
23
|
Chumakov I, Nabirotchkin S, Cholet N, Milet A, Boucard A, Toulorge D, Pereira Y, Graudens E, Traoré S, Foucquier J, Guedj M, Vial E, Callizot N, Steinschneider R, Maurice T, Bertrand V, Scart-Grès C, Hajj R, Cohen D. Combining two repurposed drugs as a promising approach for Alzheimer's disease therapy. Sci Rep 2015; 5:7608. [PMID: 25566747 PMCID: PMC5378993 DOI: 10.1038/srep07608] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/19/2014] [Indexed: 02/08/2023] Open
Abstract
Alzheimer disease (AD) represents a major medical problem where mono-therapeutic interventions demonstrated only a limited efficacy so far. We explored the possibility of developing a combinational therapy that might prevent the degradation of neuronal and endothelial structures in this disease. We argued that the distorted balance between excitatory (glutamate) and inhibitory (GABA/glycine) systems constitutes a therapeutic target for such intervention. We found that a combination of two approved drugs – acamprosate and baclofen – synergistically protected neurons and endothelial structures in vitro against amyloid-beta (Aβ) oligomers. The neuroprotective effects of these drugs were mediated by modulation of targets in GABA/glycinergic and glutamatergic pathways. In vivo, the combination alleviated cognitive deficits in the acute Aβ25–35 peptide injection model and in the mouse mutant APP transgenic model. Several patterns altered in AD were also synergistically normalised. Our results open up the possibility for a promising therapeutic approach for AD by combining repurposed drugs.
Collapse
Affiliation(s)
- Ilya Chumakov
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | - Nathalie Cholet
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Aude Milet
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Aurélie Boucard
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Damien Toulorge
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Yannick Pereira
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Esther Graudens
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Sory Traoré
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Julie Foucquier
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Mickael Guedj
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Emmanuel Vial
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | | | - Tangui Maurice
- 1] Université de Montpellier 2, 34095 Montpellier, France; Inserm, U710, 34095 Montpellier, France; EPHE, 75017 Paris, France [2] Amylgen, 2196 bd de la Lironde, 34980 Montferrier-sur-Lez, France
| | - Viviane Bertrand
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | - Rodolphe Hajj
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Daniel Cohen
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| |
Collapse
|
24
|
Faria MC, Gonçalves GS, Rocha NP, Moraes EN, Bicalho MA, Gualberto Cintra MT, Jardim de Paula J, José Ravic de Miranda LF, Clayton de Souza Ferreira A, Teixeira AL, Gomes KB, Carvalho MDG, Sousa LP. Increased plasma levels of BDNF and inflammatory markers in Alzheimer's disease. J Psychiatr Res 2014; 53:166-72. [PMID: 24576746 DOI: 10.1016/j.jpsychires.2014.01.019] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/19/2013] [Accepted: 01/30/2014] [Indexed: 01/12/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly. Neurotrophic factors and inflammatory markers may play considerable roles in AD. In this study we measured, through Enzyme-Linked Immunosorbent Assay, the plasma levels of brain derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF) and neuronal growth factor (NGF), as well as tumor necrosis factor-alpha soluble receptors, sTNFR1 and sTNFR2, and soluble intercellular adhesion molecule 1 (sICAM-1), in 50 AD patients, 37 patients with mild cognitive impairment (MCI) and 56 healthy elderly controls. BDNF levels, expressed as median and interquartile range, were higher for AD patients (2545.3, 1497.4-4153.4 pg/ml) compared to controls (1503.8, 802.3-2378.4 pg/ml), P < 0.001. sICAM-1 was also higher in AD patients. sTNFR1 levels were increased in AD when compared to controls and also to MCI. GDNF, NGF and sTNFR2 levels showed no significant differences among the studied groups. The increase in BDNF might reflect a compensatory mechanism against early neurodegeneration and seems to be related to inflammation. sTNFR1 appears to mark not only the inflammatory state but also differentiates between MCI and AD, which may be an additional tool for differentiating degrees of cognitive impairment.
Collapse
Affiliation(s)
- Mayara Chaves Faria
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av.Antonio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Gisele Santos Gonçalves
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av.Antonio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Natália Pessoa Rocha
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Edgar Nunes Moraes
- Ambulatório de Idosos do Instituto Jenny de Andrade Faria do Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Departamento de Clínica Médica da Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Aparecida Bicalho
- Ambulatório de Idosos do Instituto Jenny de Andrade Faria do Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Departamento de Clínica Médica da Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marco Túlio Gualberto Cintra
- Ambulatório de Idosos do Instituto Jenny de Andrade Faria do Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jonas Jardim de Paula
- Ambulatório de Idosos do Instituto Jenny de Andrade Faria do Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luís Felipe José Ravic de Miranda
- Ambulatório de Idosos do Instituto Jenny de Andrade Faria do Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Antônio Lúcio Teixeira
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Departamento de Clínica Médica da Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina Braga Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria das Graças Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av.Antonio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Lirlândia P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av.Antonio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
25
|
Passaro A, Dalla Nora E, Morieri ML, Soavi C, Sanz JM, Zurlo A, Fellin R, Zuliani G. Brain-derived neurotrophic factor plasma levels: relationship with dementia and diabetes in the elderly population. J Gerontol A Biol Sci Med Sci 2014; 70:294-302. [PMID: 24621946 DOI: 10.1093/gerona/glu028] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The mechanisms linking diabetes and cognitive impairment/dementia, two common conditions of elderly people, are not completely known. Brain-derived neurotrophic factor (BDNF) has antidiabetic properties, and reduced circulating BDNF was associated with dementia. We investigated the relationship between plasma BDNF levels, dementia, and diabetes in a sample of 164 community-dwelling elderly individuals, including 50 participants with vascular dementia, 44 with late onset Alzheimer's disease, 23 with cerebrovascular disease not dementia, and 47 controls (C). Presence/absence of diabetes was registered; new diagnoses of diabetes were made by the American Diabetes Association criteria. BDNF plasma levels were measured by ELISA. Both diagnosis of dementia and diabetes were associated with lower BDNF plasma values compared with the respective controls; moreover, dementia and diabetes correlated with BDNF plasma levels, independent of possible confounders. A progressive reductions of BDNF plasma levels from C (383.9 ± 204.6 pg/mL), to cerebrovascular disease not dementia (377.1 ± 130.2), to vascular dementia (313.3 ± 114.8), to late onset Alzheimer's disease (264.7 ± 147.7) was observed, (late onset Alzheimer's disease vs C, p: .03; late onset Alzheimer's disease vs cerebrovascular disease not dementia, p: .002). Demented patients affected by diabetes had the lowest BDNF mean levels (264.9 pg/mL) among individuals enrolled in this sample, suggesting the existence of a "synergistic" effect of dementia and diabetes on BDNF levels.
Collapse
Affiliation(s)
- Angela Passaro
- Department of Clinical and Experimental Medicine, Section of Internal Medicine, Gerontology and Clinical Nutrition, University of Ferrara, Italy
| | - Edoardo Dalla Nora
- Department of Clinical and Experimental Medicine, Section of Internal Medicine, Gerontology and Clinical Nutrition, University of Ferrara, Italy
| | - Mario L Morieri
- Department of Clinical and Experimental Medicine, Section of Internal Medicine, Gerontology and Clinical Nutrition, University of Ferrara, Italy
| | - Cecilia Soavi
- Department of Clinical and Experimental Medicine, Section of Internal Medicine, Gerontology and Clinical Nutrition, University of Ferrara, Italy
| | - Juana M Sanz
- Department of Clinical and Experimental Medicine, Section of Internal Medicine, Gerontology and Clinical Nutrition, University of Ferrara, Italy
| | - Amedeo Zurlo
- Operative Unit of Geriatrics, Arcispedale S. Anna, Ferrara, Italy
| | - Renato Fellin
- Department of Clinical and Experimental Medicine, Section of Internal Medicine, Gerontology and Clinical Nutrition, University of Ferrara, Italy
| | - Giovanni Zuliani
- Department of Clinical and Experimental Medicine, Section of Internal Medicine, Gerontology and Clinical Nutrition, University of Ferrara, Italy.
| |
Collapse
|
26
|
Lin Y, Yao J, Chen Y, Pang L, Li H, Cao Z, You K, Dai H, Wu R. Hippocampal neurochemical changes in senescent mice induced with chronic injection of D-galactose and NaNO₂: an in vitro high-resolution NMR spectroscopy study at 9.4T. PLoS One 2014; 9:e88562. [PMID: 24533108 PMCID: PMC3922890 DOI: 10.1371/journal.pone.0088562] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 01/13/2014] [Indexed: 02/05/2023] Open
Abstract
Proton magnetic resonance spectroscopy (¹H-MRS) has been used to provide useful information about the neurochemical changes reflecting early pathological alterations in Alzheimer's disease (AD) brain. In this study, we have longitudinally measured the hippocampal neurochemical profile in vitro in senescent mice induced with chronic injection of D-Galactose and NaNO₂, at different time point from day 30 to day 70 with a 10-day interval. Pathological brain alterations induced by D-Galactose and NaNO₂ were monitored through hematoxylin and eosin (HE) staining, Congo red staining and bielschowsky silver staining, and the cognition deficits were assessed via Morris Water Maze (MWM) test. This D-galactose and NaNO₂ treated mouse model, characterized by an early-onset memory dysfunction, a robust neuronal loss, amyloid plaques and neurofibrillary tangles in hippocampal subdivision, well mimics a prodromal Alzheimer's phenotype. Consistent with previously published in vivo ¹H MRS findings in human AD patients and AD transgenic mice, our in vitro ¹H MRS on the perchloric acid extractions of hippocampus in senescent mice observed significant decreases of N-acetylaspartate (NAA) and Glutamate (Glu) but an increase in Myo-inositol (mIns). Elevated mIns occurred prior to the reduction of NAA and Glu during the progression of aging. In addition, changes in mIns, NAA and Glu were found to precede pathological abnormalities. Overall, our in vitro findings in senescent mice validated the concept that hippocampal neurochemical alternations preceded the pathological changes of the brain, and could serve as potential markers of AD progression. Reductions of NAA and Glu can be interpreted in terms of neuronal degeneration and dysfunctions in glutamatergic activity that may contribute to the pathophysiological mechanisms underlying AD. Elevated mIns might be related to glial activation. Further experiments are needed to explore the potential value of mIns in the early diagnosis of AD, to verify whether glial cell proliferation occurs earlier than neuronal changes.
Collapse
Affiliation(s)
- Yan Lin
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Jianli Yao
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China
- Sichuan Provincial Tumor Hospital, Chengdu, China
| | - Yaowen Chen
- Shantou University Central Laboratory and NMR Unit, Shantou, Guangdong Province, China
| | - Li Pang
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Haihong Li
- Mental Health Center of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Zhen Cao
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Kezeng You
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Haiyang Dai
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Renhua Wu
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China
- Key Laboratory of Molecular Imaging of Guangdong Province, Shantou, China
- * E-mail:
| |
Collapse
|
27
|
Wang Y, Yin H, Wang L, Shuboy A, Lou J, Han B, Zhang X, Li J. Curcumin as a potential treatment for Alzheimer's disease: a study of the effects of curcumin on hippocampal expression of glial fibrillary acidic protein. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2013; 41:59-70. [PMID: 23336507 DOI: 10.1142/s0192415x13500055] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Curcumin, an agent traditionally utilized for its preventative action against tumorigenesis, oxidation, inflammation, apoptosis and hyperlipemia, has also been used in the treatment of Alzheimer's disease (AD). Recent advances in the study of AD have revealed astrocytes (AS) as being key factors in the early pathophysiological changes in AD. Glial fibrillary acidic protein (GFAP), a marker specific to AS, is markedly more manifest during morphological modifications and neural degeneration signature during the onset of AD. Several studies investigating the functionality of curcumin have shown that it not only inhibits amyloid sedimentation but also accelerates the disaggregation of amyloid plaque. Thus, we are interested in the relationship between curcumin and spatial memory in AD. In this study, we intend to investigate the effects of curcumin in amyloid-β (Aβ(1-40)) induced AD rat models on both the behavioral and molecular levels, that is to say, on their spatial memory and on the expression of GFAP in their hippocampi. Our results were statistically significant, showing that the spatial memory of AD rats improved following curcumin treatment (p < 0.05), and that the expression of GFAP mRNA and the number of GFAP positive cells in the curcumin treated rats was decreased relative to the AD group rats (p < 0.05). Furthermore, the expression level of GFAP mRNA in hippocampal AS in the AD rats significantly increased when compared with that in the sham control (p < 0.05). Taken together, these results suggest that curcumin improves the spatial memory disorders (such disorders being symptomatic of AD) in Aβ(1-40)-induced rats by down regulating GFAP expression and suppressing AS activity.
Collapse
Affiliation(s)
- Yunliang Wang
- Department of Neurology, The 148th Hospital, Zibo, Shandong 255300, China.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Crish SD, Dapper JD, MacNamee SE, Balaram P, Sidorova TN, Lambert WS, Calkins DJ. Failure of axonal transport induces a spatially coincident increase in astrocyte BDNF prior to synapse loss in a central target. Neuroscience 2012; 229:55-70. [PMID: 23159315 DOI: 10.1016/j.neuroscience.2012.10.069] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 10/04/2012] [Accepted: 10/24/2012] [Indexed: 01/07/2023]
Abstract
Failure of anterograde transport to distal targets in the brain is a common feature of neurodegenerative diseases. We have demonstrated in rodent models of glaucoma, the most common optic neuropathy, early loss of anterograde transport along the retinal ganglion cell (RGC) projection to the superior colliculus (SC) is retinotopic and followed by a period of persistence of RGC axon terminals and synapses through unknown molecular pathways. Here we use the DBA/2J mouse model of hereditary glaucoma and an acute rat model to demonstrate that retinotopically focal transport deficits in the SC are accompanied by a spatially coincident increase in brain-derived neurotrophic factor (BDNF), especially in hypertrophic astrocytes. These neurochemical changes occur prior to loss of RGC synapses in the DBA/2J SC. In contrast to BDNF protein, levels of Bdnf mRNA decreased with transport failure, even as mRNA encoding synaptic structures remained unchanged. In situ hybridization signal for Bdnf mRNA was the strongest in SC neurons, and labeling for the immature precursor pro-BDNF was very limited. Subcellular fractionation of SC indicated that membrane-bound BDNF decreased with age in the DBA/2J, while BDNF released from vesicles remained high. These results suggest that in response to diminished axonal function, activated astrocytes in the brain may sequester mature BDNF released from target neurons to counter stressors that otherwise would challenge survival of projection synapses.
Collapse
Affiliation(s)
- S D Crish
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, 11425 Langford Medical Research Building IV, 2213 Garland Avenue, Nashville, TN 37232, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Zhang F, Lu YF, Wu Q, Liu J, Shi JS. Resveratrol promotes neurotrophic factor release from astroglia. Exp Biol Med (Maywood) 2012; 237:943-8. [DOI: 10.1258/ebm.2012.012044] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Neurotrophic factors such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) are considered to contribute to the development, maintenance and survival of neurons, glia and oligodendrocytes. Astroglia are a major source of various neurotrophic factors. Thus, enhancement of astroglia-mediated neurotrophic factor release might hold promising potential for neurological diseases. Resveratrol, a natural non-flavonoid polyphenol found in grapes and red wine, has been recognized to be beneficial for health. Here, rat primary astroglia-enriched cultures were used to investigate the effects of resveratrol-mediated neurotrophic factor release and the related mechanisms. The cultures were treated with 25–100 μmmol/L resveratrol for 12–48 h. Results showed resveratrol increased BDNF and GDNF production in the culture medium. In addition, the production of BDNF in the supernatant of cultures was increased five-fold over control cultures 24 h after resveratrol treatment and then remained high 36 h later. Meanwhile, the production of GDNF was initially increased by up to four-fold 24 h after resveratrol treatment and continued to increase to six-fold at 36 h and remained at a high level till 48 h. Western blot analysis of BDNF and GDNF protein in astroglia at different time points after resveratrol treatment indicated similar increases. Furthermore, resveratrol significantly induced the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and cAMP responsive element-binding protein (CREB) in astroglia. Overall, resveratrol is effective in promoting astroglia-derived neurotrophic factor release, and this effect is mediated, at least in part, by the activation of ERK1/2 and CREB.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Pharmacology and Key Lab of Basic Pharmacology of Guizhou, Zunyi Medical College, 563099 Zunyi, People's Republic of China
| | - Yuan-Fu Lu
- Department of Pharmacology and Key Lab of Basic Pharmacology of Guizhou, Zunyi Medical College, 563099 Zunyi, People's Republic of China
| | - Qin Wu
- Department of Pharmacology and Key Lab of Basic Pharmacology of Guizhou, Zunyi Medical College, 563099 Zunyi, People's Republic of China
| | - Jie Liu
- University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jing-Shan Shi
- Department of Pharmacology and Key Lab of Basic Pharmacology of Guizhou, Zunyi Medical College, 563099 Zunyi, People's Republic of China
| |
Collapse
|
31
|
Chen SQ, Cai Q, Shen YY, Wang PJ, Teng GJ, Zhang W, Zang FC. Age-related changes in brain metabolites and cognitive function in APP/PS1 transgenic mice. Behav Brain Res 2012; 235:1-6. [PMID: 22828014 DOI: 10.1016/j.bbr.2012.07.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 07/08/2012] [Accepted: 07/10/2012] [Indexed: 12/23/2022]
Abstract
Proton magnetic resonance spectroscopy ((1)H-MRS) and the Morris water maze (MWM) have played an important role in Alzheimer's disease (AD) research. The aim of this study was to determine whether (1)H-MRS and the MWM can detect for early AD in APP/PS1 transgenic (tg) mice. (1)H-MRS was performed in 20 tg mice and 15 wild-type mice at 3, 5 and 8 months of age. The concentration of N-acetylaspartate (NAA), glutamate (Glu), myo-inositol (mI), choline (Cho) and creatine (Cr) in the hippocampus were measured, and the NAA/Cr, Glu/Cr, mI/Cr and Cho/Cr ratios were quantified. Additionally, the spatial learning and memory of the mice were evaluated by MWM. The (1)H-MRS revealed that mI levels in tg mice were significantly higher at 3 months of age compared to wt mice, while the NAA and Glu levels in 5- and 8-month-old tg mice were significantly decreased (p<0.05). Additionally, significant cognitive changes only occurred at 8 months of age in APP/PS1 tg mice. These results indicated that metabolic changes preceded overt cognitive dysfunctions in early-stage AD, suggesting that (1)H-MRS is a more sensitive biomarker for assessing early AD.
Collapse
Affiliation(s)
- Shuang-qing Chen
- Department of Radiology, Suzhou Hospital, Nanjing Medical University, Suzhou 215001, China.
| | | | | | | | | | | | | |
Collapse
|
32
|
Iwasaki Y, Negishi T, Inoue M, Tashiro T, Tabira T, Kimura N. Sendai virus vector-mediated brain-derived neurotrophic factor expression ameliorates memory deficits and synaptic degeneration in a transgenic mouse model of Alzheimer's disease. J Neurosci Res 2012; 90:981-9. [DOI: 10.1002/jnr.22830] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/18/2011] [Accepted: 10/21/2011] [Indexed: 11/11/2022]
|
33
|
Kimura N, Okabayashi S, Ono F. Dynein dysfunction disrupts intracellular vesicle trafficking bidirectionally and perturbs synaptic vesicle docking via endocytic disturbances a potential mechanism underlying age-dependent impairment of cognitive function. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:550-61. [PMID: 22182700 DOI: 10.1016/j.ajpath.2011.10.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/18/2011] [Accepted: 10/25/2011] [Indexed: 01/09/2023]
Abstract
Although genetic studies have demonstrated that β-amyloid protein (Aβ) plays a pivotal role in Alzheimer's disease (AD) pathogenesis, how aging contributes to AD onset remains unclear. Moreover, growing evidence suggests that Aβ-independent mechanisms, such as altered intracellular signaling cascades and impaired neurotransmitter release, also are likely involved in this process. Cytoplasmic dynein, a microtubule-based motor protein, mediates minus end-directed vesicle transport via interactions with dynactin, another microtubule-associated protein. We previously showed that normal aging attenuates the interaction between dynein-dynactin complexes in monkey brain and that dynein dysfunction reproduces age-dependent endocytic disturbances, resulting in intracellular Aβ accumulation. In this study, we report that dynein dysfunction disrupts not only retrograde transport of neurotrophic receptors but also anterograde transport of synaptic vesicles, which occurs concomitantly with an increase in Rab3 GTPase levels. Additionally, synaptic vesicle docking was perturbed via enhanced endocytosis. Dynein dysfunction also induced neuritic swelling, which is accompanied by a significant accumulation of neurofilaments. Moreover, we also confirmed that the dynein dysfunction-related disturbances are associated with aging in monkey brains and that age-dependent endocytic disturbances precede Aβ abnormality. These findings suggest that dynein dysfunction can alter neuronal activity via endocytic disturbances and may underlie age-dependent impairment of cognitive function. Moreover, in the presence of other risk factors, such as intracellular Aβ accumulation, dynein dysfunction may contribute to the development of AD.
Collapse
Affiliation(s)
- Nobuyuki Kimura
- Laboratory of Disease Control, Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Ibaraki, Japan.
| | | | | |
Collapse
|
34
|
Diniz BS, Teixeira AL. Brain-derived neurotrophic factor and Alzheimer's disease: physiopathology and beyond. Neuromolecular Med 2011; 13:217-22. [PMID: 21898045 DOI: 10.1007/s12017-011-8154-x] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Accepted: 08/26/2011] [Indexed: 01/25/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is the most widely distributed neurotrophin in the central nervous system where it plays several pivotal roles in synaptic plasticity and neuronal survival. As a consequence, BDNF became a key target in the physiopathology of several neurological and psychiatric diseases. Recent studies have reported altered levels of BDNF in the circulation, i.e. serum or plasma, of patients with Alzheimer's disease (AD), and low BDNF levels in the CSF as predictor of future cognitive decline in healthy older subjects. Altered BDNF circulating levels have also been reported in other neurodegenerative and psychiatric disorders, hampering its use as a specific biomarker for AD. Therefore, BDNF seems to be an unspecific biomarker of neuropsychiatric disorders marked by neurodegenerative changes.
Collapse
Affiliation(s)
- Breno Satler Diniz
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, Rua Dr. Ovidio Pires de Campos, 785 3rd floor, São Paulo, SP, 05403-010, Brazil.
| | | |
Collapse
|
35
|
Hou L, Liu Y, Wang X, Ma H, He J, Zhang Y, Yu C, Guan W, Ma Y. The effects of amyloid-β42 oligomer on the proliferation and activation of astrocytes in vitro. In Vitro Cell Dev Biol Anim 2011; 47:573-80. [DOI: 10.1007/s11626-011-9439-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 06/24/2011] [Indexed: 01/14/2023]
|
36
|
Teixeira AL, Barbosa IG, Diniz BS, Kummer A. Circulating levels of brain-derived neurotrophic factor: correlation with mood, cognition and motor function. Biomark Med 2011; 4:871-87. [PMID: 21133708 DOI: 10.2217/bmm.10.111] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is the most widely distributed neurotrophin in the CNS, where it plays several pivotal roles in synaptic plasticity and neuronal survival. As a consequence, BDNF has become a key target in the physiopathology of several neurological and psychiatric diseases. Recent studies have consistently reported altered levels of BDNF in the circulation (i.e., serum or plasma) of patients with major depression, bipolar disorder, Alzheimer's disease, Huntington's disease and Parkinson's disease. Correlations between serum BDNF levels and affective, cognitive and motor symptoms have also been described. BDNF appears to be an unspecific biomarker of neuropsychiatric disorders characterized by neurodegenerative changes.
Collapse
Affiliation(s)
- Antonio Lucio Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica & Imunologia, Instituto de Ciências Biológicas, UFMG Avenue Antonio Carlos, 6627 - 31270-901 - Belo Horizonte, MG, Brazil.
| | | | | | | |
Collapse
|
37
|
Figueiró M, Ilha J, Linck VM, Herrmann AP, Nardin P, Menezes CB, Achaval M, Gonçalves CA, Porciúncula LO, Nunes DS, Elisabetsky E. The Amazonian herbal Marapuama attenuates cognitive impairment and neuroglial degeneration in a mouse Alzheimer model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2011; 18:327-333. [PMID: 20739160 DOI: 10.1016/j.phymed.2010.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
UNLABELLED Alzheimer's disease (AD) is expected to affect more than 22 million people worldwide by 2025, causing devastating suffering and enormous costs to families and society. AD is a multifactorial disease, with a complex pathological mosaic. In rodents, AD-like dementia can be induced by cerebral microinjection of Aβ peptide, leading to amyloid deposits, amnesia and various features of neurodegeneration. Marapuama (Ptychopetalum olacoides) is regarded as a "brain tonic" in the Amazon region and shows a nootropic profile in rodents. AIM OF THE STUDY Because a specific extract (POEE) of Marapuama was shown to possess promnesic and anti-amnesic properties, the aim of this study was to verify if POEE is also effective against Aβ(1-42)-induced cognitive deficit in mice. Additionally, Aβ deposits (Congo red), GFAP immunoreactivity (immunohistochemistry), and neurodegenerative changes in the hippocampal pyramidal layer (Nissl) were examined as measures of Aβ(1-42)-induced neurodegeneration. MATERIALS AND METHODS CF1 mice were subjected to the experimental Alzheimer model with the Aβ(1-42) i.c.v. administration. The effects of POEE 800 mg/kg were evaluated over 14 consecutive days of treatment. RESULTS The data show that 14 days of oral treatment with POEE (800 mg/kg) was effective in preventing Aβ-induced cognitive impairment, without altering the levels of BDNF and with parallel reductions in Aβ deposits and astrogliosis. CA1 hippocampus loss induced by Aβ(1-42) was also diminished in POEE-treated mice. CONCLUSION This study offers evidence of functional and neuroprotective effects of two weeks treatment with a Ptychopetalum olacoides extract against Aβ peptide-induced neurotoxicity in mice. Given the multifactorial nature of neurodegeneration, the considerable potential for an AChE inhibitor displaying associated neuroprotective properties such as here reported warrants further clinic evaluation.
Collapse
Affiliation(s)
- M Figueiró
- Laboratório de Etnofarmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Porto Alegre, RS, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Strain-specific BDNF expression of rat primary astrocytes. J Neuroimmunol 2010; 220:90-8. [PMID: 20176405 DOI: 10.1016/j.jneuroim.2010.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 02/01/2010] [Accepted: 02/02/2010] [Indexed: 12/15/2022]
Abstract
By producing brain-derived neurotrophic factor (BDNF), astrocytes play a role in disease resistance. This study was undertaken to investigate whether primary astrocytes derived from LEW/N and F344/N rats differentially express BDNF. LEW/N astrocytes expressed more BDNF mRNA and protein than F344/N astrocytes in basal and valproic acid (VPA)-stimulated conditions. VPA suppresses HDAC enzyme activity without affecting HDAC gene and protein expression in astrocytes of both strains. Blockade of TrkB receptors resulted in similar fold decreases in basal BDNF mRNA levels between two strains. The results suggest that inhibition of HDAC activity and BDNF-TrkB autocrine loop are involved in regulation of astrocytic BDNF transcription, whereas the mechanisms for elevated constitutive gene BDNF expression of LEW/N astrocytes remain to be investigated.
Collapse
|
39
|
Chen SQ, Wang PJ, Ten GJ, Zhan W, Li MH, Zang FC. Role of myo-inositol by magnetic resonance spectroscopy in early diagnosis of Alzheimer's disease in APP/PS1 transgenic mice. Dement Geriatr Cogn Disord 2010; 28:558-66. [PMID: 20093832 PMCID: PMC2837893 DOI: 10.1159/000261646] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS To explore the potential value of myo-inositol (mIns), which is regarded as a biomarker for early diagnosis of Alzheimer's disease, in APP/PS1 transgenic (tg) mice detected by (1)H-MRS. METHODS (1)H-MRS was performed in 30 APP/PS1 tg mice and 20 wild-type (wt) littermates at 3, 5 and 8 months of age. Areas under the peak of N-acetylaspartate (NAA), mIns and creatine (Cr) in the frontal cortex and hippocampus were measured, and the NAA/Cr and mIns/Cr ratios were analyzed quantitatively. RESULTS Compared with the wt mice, the mIns/Cr ratio of the 3-month-old tg mice was significantly higher (p < 0.05), and pathology showed activation and proliferation of astrocytes in the frontal cortex and hippocampus. The concentration of NAA was significantly lower at 8 and 8 months of age (p < 0.05). According to the threshold of mIns/Cr that was adopted to separate the tg from the wt mice, the rate of correct predictions was 82, 94 and 95%, respectively, for 3, 5 and 8 months. CONCLUSION Of the early AD metabolites as detected by (1)H-MRS, mIns is the most valuable marker for assessment of AD. Quantitative analysis of mIns may provide important clues for early diagnosis of AD.
Collapse
Affiliation(s)
- Shuang-Qing Chen
- Department of Radiology, Tongji Hospital, Tongji University, Shanghai, China
| | - Pei-Jun Wang
- Department of Radiology, Tongji Hospital, Tongji University, Shanghai, China,*Pei-Jun Wang, Department of Radiology, Tongji Hospital, Tongji University, No. 389, Xincun Road, Putuo District, Shanghai 200065 (China), Tel. +86 21 6611 1206, Fax +86 21 5695 2231, E-Mail
| | - Gao-Jun Ten
- Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China,Molecular Imaging Laboratory, Zhongda Hospital, Southeast University, Nanjing, China
| | - Wei Zhan
- Department of Radiology, Tongji Hospital, Tongji University, Shanghai, China
| | - Ming-Hua Li
- Department of Radiology, Tongji Hospital, Tongji University, Shanghai, China
| | - Feng-Chao Zang
- Molecular Imaging Laboratory, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
40
|
Li Y, Li T, Zhuang M, Wang K, Zhang J, Shi N. High-dose dibutyl phthalate improves performance of F1 generation male rats in spatial learning and increases hippocampal BDNF expression independent on p-CREB immunocontent. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2010; 29:32-38. [PMID: 21787579 DOI: 10.1016/j.etap.2009.09.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 07/15/2009] [Accepted: 09/02/2009] [Indexed: 05/31/2023]
Abstract
Dibutyl phthalate (DBP), an important representative of endocrine disrupting chemical, is suspected of affecting the cognitive function of humans and animals. In this study, effects of DBP on maze performance in male rats were evaluated by spatial learning tasks; the effects of DBP on the expression of brain-derived neurotrophic factor (BDNF) were also analyzed in both mRNA and mature protein levels in the hippocampus, with intent to investigate the possible mechanism underlying the behavioral findings. Pregnant Wistar rats were treated orally by gavage with 0, 25, 75, 225 and 675mgDBP/kgBW/day from gestational day (GD) 6 to postnatal day (PND) 21, and then the weaned offspring continued receiving the same treatment till PND 28. We found that male pups treated with high-dose DBP showed enhancement in spatial acquisition in a Morris water maze during PNDs 30-33, and displayed better retention of spatial memory in a probe trial after a reverse trail during PNDs 60-62. Real-time PCR and western blotting analysis of the hippocampus from DBP-treated male rats on PND 21 revealed an increase in BDNF expression, compared to the vehicle-matched control. BDNF variant III, a transcription promoted by active CREB (i.e. p-CREB), as well as the immunocontent of p-CREB, was scarcely altered by the treatment. Our results suggest that developmental treatment with high-dose DBP improves spatial memory in male rats, and this effect may be related to an increase in BDNF expression in the hippocampus in a p-CREB independent route.
Collapse
Affiliation(s)
- Yuanfeng Li
- Department of Health Toxicology, MOE Key Laboratory of Environmental and Health, Tongji Medical College of Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan 430030, Hubei, China
| | | | | | | | | | | |
Collapse
|
41
|
Aliaga E, Silhol M, Bonneau N, Maurice T, Arancibia S, Tapia-Arancibia L. Dual response of BDNF to sublethal concentrations of beta-amyloid peptides in cultured cortical neurons. Neurobiol Dis 2009; 37:208-17. [PMID: 19822210 DOI: 10.1016/j.nbd.2009.10.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 09/25/2009] [Accepted: 10/02/2009] [Indexed: 02/06/2023] Open
Abstract
Beta-amyloid (Abeta) deposition is one important pathological hallmark in Alzheimer's disease (AD). However, low levels of Abeta may modify critical endogenous protection systems before neurodegeneration occurs. We examined the time-course effect of sublethal concentrations of Abeta on total BDNF (panBDNF), BDNF transcripts (I, II, IV and VI), trkB.FL, trkB.T1 and p75(NGFR) mRNA expression in cultured cortical neurons. We have shown that Abeta exhibited a dual response on BDNF mRNA, i.e. an increase at short times (3-5 h) and a dramatic decrease at longer times (24 or 48 h). The early increase in BDNF expression seems to be driven by increased expression of transcripts I and IV. The BDNF drop was specific since did not occur for other mRNAs examined. The BDNF protein content showed a similar profile but did not follow the dramatic reduction as its encoding mRNA. These observations may help to explain cognitive deficits observed at initial stages of AD.
Collapse
Affiliation(s)
- E Aliaga
- Centro de Neurobiología y Plasticidad del Desarrollo, Departamento de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña 1111, Playa Ancha, Valparaíso, Chile
| | | | | | | | | | | |
Collapse
|
42
|
Gunstad J, Benitez A, Smith J, Glickman E, Spitznagel MB, Alexander T, Juvancic-Heltzel J, Murray L. Serum brain-derived neurotrophic factor is associated with cognitive function in healthy older adults. J Geriatr Psychiatry Neurol 2008; 21:166-70. [PMID: 18503034 DOI: 10.1177/0891988708316860] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cognitive decline is common in older adults, even in the absence of significant medical or neurological conditions. Recent work implicates serum levels of brain-derived neurotrophic factor in age-related cognitive decline, though no study has directly examined this possibility. A total of 35 older adults without neurological history underwent fasting blood draw and completed a brief neuropsychological test battery during a single session. After adjusting for demographic and medical confounds, higher serum brain-derived neurotrophic factor levels were associated with better performance on the Mini-Mental State Examination (r = .36) and short form of the Boston Naming Test (r = .39). These findings extend work from Alzheimer disease and vascular dementia samples and indicate that higher brain-derived neurotrophic factor levels are associated with better neuropsychological function in healthy older adults. The exact mechanisms for this relationship are unknown and require further examination.
Collapse
Affiliation(s)
- John Gunstad
- Department of Psychology, Kent State University, Kent, OH 44242, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Lanctôt KL, Herrmann N, Rothenburg LS. Commentary: Aβ(1–42) injection causes memory impairment, lowered cortical and serum BDNF levels, and decreased hippocampal 5-HT2A levels. Exp Neurol 2008; 212:251-4. [DOI: 10.1016/j.expneurol.2008.04.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Revised: 03/24/2008] [Accepted: 04/28/2008] [Indexed: 12/20/2022]
|
44
|
Liu RY, Gu R, Qi XL, Zhang T, Zhao Y, He Y, Pei JJ, Guan ZZ. Decreased nicotinic receptors and cognitive deficit in rats intracerebroventricularly injected with beta-amyloid peptide(1-42) and fed a high-cholesterol diet. J Neurosci Res 2008; 86:183-93. [PMID: 17705292 DOI: 10.1002/jnr.21463] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To investigate whether the changes in nicotinic receptors (nAChRs) and in learning and memory associated with Alzheimer's disease (AD) are influenced by both beta-amyloid peptide (Abeta) and cholesterol in vivo, we examined the effects of intracerebroventricular injection of Abeta(1-42) and/or a high-cholesterol diet on brain levels of nAChRs and learning and memory in rats. The levels of nAChR subunit proteins and the corresponding mRNA were measured by Western blotting and RT-PCR, respectively; and learning and memory were evaluated with the Morris Water Maze examination. Injection of Abeta(1-42) resulted in deposition of this peptide, activation of astrocytes, decreased levels of the alpha7 and alpha4 protein subunits of the nAChR, and elevated expression of alpha7 mRNA, as well as impaired learning and spatial memory. A high-cholesterol diet activated astrocytes and, more importantly, potentiated the toxic effects of Abeta on nAChR subunit levels and on learning and memory. These findings may be highly relevant to the mechanisms underlying the cognitive deficits associated with AD.
Collapse
Affiliation(s)
- Ru-Yu Liu
- Department of Pathology, Guiyang Medical University, Guizhou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Taylor AR, Gifondorwa DJ, Newbern JM, Robinson MB, Strupe JL, Prevette D, Oppenheim RW, Milligan CE. Astrocyte and muscle-derived secreted factors differentially regulate motoneuron survival. J Neurosci 2007; 27:634-44. [PMID: 17234595 PMCID: PMC6672790 DOI: 10.1523/jneurosci.4947-06.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Revised: 11/14/2006] [Accepted: 12/11/2006] [Indexed: 01/03/2023] Open
Abstract
During development, motoneurons (MNs) undergo a highly stereotyped, temporally and spatially defined period of programmed cell death (PCD), the result of which is the loss of 40-50% of the original neuronal population. Those MNs that survive are thought to reflect the successful acquisition of limiting amounts of trophic factors from the target. In contrast, maturation of MNs limits the need for target-derived trophic factors, because axotomy of these neurons in adulthood results in minimal neuronal loss. It is unclear whether MNs lose their need for trophic factors altogether or whether, instead, they come to rely on other cell types for nourishment. Astrocytes are known to supply trophic factors to a variety of neuronal populations and thus may nourish MNs in the absence of target-derived factors. We investigated the survival-promoting activities of muscle- and astrocyte-derived secreted factors and found that astrocyte-conditioned media (ACM) was able to save substantially more motoneurons in vitro than muscle-conditioned media (MCM). Our results indicate that both ACM and MCM are significant sources of MN trophic support in vitro and in ovo, but only ACM can rescue MNs after unilateral limb bud removal. Furthermore, we provide evidence suggesting that MCM facilitates the death of a subpopulation of MNs in a p75(NTR) - and caspase-dependent manner; however, maturation in ACM results in MN trophic independence and reduced vulnerability to this negative, pro-apoptotic influence from the target.
Collapse
Affiliation(s)
- Anna R Taylor
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|