1
|
Bulgay C, Zorba E, Kazan HH, Bayraktar I, Uca M, Ergün MA, John G, Yusupov RA, Sultanov RI, Semenova EA, Larin AK, Kulemin NA, Generozov EV, Ahmetov II. BDNF coexpresses with MTOR and is associated with muscle fiber size, lean mass and power-related traits. Eur J Appl Physiol 2025:10.1007/s00421-025-05804-3. [PMID: 40299026 DOI: 10.1007/s00421-025-05804-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE Recent research suggests a link between brain-derived neurotrophic factor (BDNF) and the mTOR signaling pathway, a key regulator of protein synthesis and muscle growth. However, it remains unclear whether BDNF influences muscle growth and power performance. Our study aimed to investigate the relationship between the expression of BDNF and MTOR genes in human skeletal muscle and examine the association between genetically predicted higher expression of the BDNF gene and muscle fiber size, lean mass, power performance, and power athlete status. METHODS The study involved 456,382 subjects, including 285 athletes, 112 physically active individuals with muscle fiber composition data, 291 sedentary individuals with gene expression data, 5451 controls, and 450,243 UK Biobank participants. The muscle fiber composition was evaluated using immunohistochemistry, while gene expression analysis was performed using RNA sequencing. BDNF genotyping was carried out using real-time PCR or microarrays. RESULTS We found that BDNF gene expression was positively associated with MTOR gene expression in the vastus lateralis (p < 0.0001). Furthermore, genetically predicted higher BDNF expression (i.e., carriage of the C allele of the rs6265 (Val66Met) BDNF polymorphism) was positively associated with the cross-sectional area of fast-twitch muscle fibers in athletes (p = 0.0069), appendicular lean mass (p = 2.6 × 10⁻⁷), personal best scores of power athletes (p = 0.029), and power athlete status (p = 0.0056). CONCLUSION Our study demonstrates a positive correlation between BDNF and MTOR gene expression in human skeletal muscle, with genetically predicted higher BDNF expression associated with greater muscle fiber size, lean mass, enhanced power performance, and power athlete status.
Collapse
Affiliation(s)
- Celal Bulgay
- Sports Science Faculty, Bingol University, Bingol, Türkiye
| | - Erdal Zorba
- Sport Sciences Faculty, Gazi University, Ankara, Türkiye
| | - Hasan H Kazan
- Department of Medical Biology, Gulhane Faculty of Medicine, University of Health Sciences, Ankara, Türkiye
| | - Işık Bayraktar
- Sports Science Faculty, Alanya Alaaddin Keykubat University, Alanya, Türkiye
| | - Merve Uca
- School of Physical Education and Sports, İstanbul Esenyurt University, Istanbul, Türkiye
| | - Mehmet A Ergün
- Department of Medical Genetics, Faculty of Medicine, Gazi University, Ankara, Türkiye
| | - George John
- Transform Specialist Medical Centre, Dubai, United Arab Emirates
| | - Rinat A Yusupov
- Department of Physical Culture and Sport, Kazan National Research Technical University Named After A.N. Tupolev-KAI, Kazan, Russia
| | - Rinat I Sultanov
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Ekaterina A Semenova
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Research Institute of Physical Culture and Sport, Volga Region State University of Physical Culture, Sport and Tourism, Kazan, Russia
| | - Andrey K Larin
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Nikolay A Kulemin
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Edward V Generozov
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Ildus I Ahmetov
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.
- Sports Genetics Laboratory, St Petersburg Research Institute of Physical Culture, St Petersburg, Russia.
- Laboratory of Genetics of Aging and Longevity, Kazan State Medical University, Kazan, Russia.
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
2
|
Tan Z, Ping J, Zhang Y, Kong C, Luo J, Liu X. The impact of the interaction between BDNF rs7103411 gene polymorphism and social activities on mild cognitive impairment in community-dwelling elderly adults. Front Psychiatry 2025; 15:1469671. [PMID: 39876995 PMCID: PMC11772356 DOI: 10.3389/fpsyt.2024.1469671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/12/2024] [Indexed: 01/31/2025] Open
Abstract
Objective To investigate the correlation between BDNF gene polymorphism, BDNF levels, and susceptibility to mild cognitive impairment (MCI). Methods In this study, we investigated 107 elderly adults individuals from a community in Zhongshan, Guangdong Province, with an average age of 73.17 ± 7.081 years. The participants included 52 patients with Mild Cognitive Impairment due to Alzheimer's Disease and 55 cognitively normal elderly adults control subjects. The two groups were matched based on gender, age, and education level. We assessed their cognitive functions and analyzed their genotypes and serum BDNF levels. Analysis of covariance (ANCOVA) was used to evaluate the differences in serum BDNF levels between the MCI group and the control group. Multivariate linear regression was utilized to analyze the association between BDNF levels and susceptibility to MCI, as well as cognitive functions. Multivariate logistic regression was employed to investigate the association between BDNF gene polymorphisms and the risk of developing MCI, along with their interactions. Results The ANCOVA analysis indicated that there was no significant difference in serum BDNF levels between the MCI group and the control group (P > 0.05). Correlation analysis revealed a negative correlation between Mini-Mental Status Examination (MMSE) total scores and MCI (r = -0.461, P = 0.001), with significant correlations observed in orientation (r = -0.420, P = 0.002). Multiple linear regression analysis showed that specific polymorphisms, including rs7103411 (CT+TT vs. CC), rs6265 (CT and CT+TT vs. CC), rs11030104 (AG and AG+GG vs. AA), and rs988748 (CG+CC vs. GG), were significantly associated with decreased serum BDNF levels (P < 0.05). Multivariate logistic regression showed that rs7103411 polymorphism was associated with susceptibility to MCI; individuals with the CT or CC genotype had a 0.370 times lower risk of developing MCI compared to those with the TT genotype (OR = 0.370, 95% CI: 0.141-0.970, P = 0.043). A significant interaction was found between rs7103411 and social activity, which influenced the risk of developing MCI. Specifically, individuals with the CT or TT genotype of rs7103411 who engaged in social activities had a significantly lower risk of developing MCI (OR = 0.32, 95% CI: 0.117-0.878, P = 0.027). Conclusion This study indicates that BDNF rs7103411、rs6265、rs11030104 and rs988748 are associated with decreased serum BDNF levels in MCI patients. Individuals carrying the TT genotype in the BDNF rs7103411 gene are associated with an increased susceptibility to MCI. Individuals with the rs7103411 CT or TT genotype who participated in social activities showed a significantly reduced risk of developing MCI, suggesting that the interaction between the BDNF rs7103411 genotype and social activity can help reduce the risk of MCI.
Collapse
Affiliation(s)
| | | | | | | | | | - Xinxia Liu
- Department of Psychiatry, Third People’s Hospital of Zhongshan City, Zhongshan, China
| |
Collapse
|
3
|
Vázquez-Lizarraga R, Mendoza-Viveros L, Cid-Castro C, Ruiz-Montoya S, Carreño-Vázquez E, Orozco-Solis R. Hypothalamic circuits and aging: keeping the circadian clock updated. Neural Regen Res 2024; 19:1919-1928. [PMID: 38227516 PMCID: PMC11040316 DOI: 10.4103/1673-5374.389624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/22/2023] [Accepted: 10/20/2023] [Indexed: 01/17/2024] Open
Abstract
Over the past century, age-related diseases, such as cancer, type-2 diabetes, obesity, and mental illness, have shown a significant increase, negatively impacting overall quality of life. Studies on aged animal models have unveiled a progressive discoordination at multiple regulatory levels, including transcriptional, translational, and post-translational processes, resulting from cellular stress and circadian derangements. The circadian clock emerges as a key regulator, sustaining physiological homeostasis and promoting healthy aging through timely molecular coordination of pivotal cellular processes, such as stem-cell function, cellular stress responses, and inter-tissue communication, which become disrupted during aging. Given the crucial role of hypothalamic circuits in regulating organismal physiology, metabolic control, sleep homeostasis, and circadian rhythms, and their dependence on these processes, strategies aimed at enhancing hypothalamic and circadian function, including pharmacological and non-pharmacological approaches, offer systemic benefits for healthy aging. Intranasal brain-directed drug administration represents a promising avenue for effectively targeting specific brain regions, like the hypothalamus, while reducing side effects associated with systemic drug delivery, thereby presenting new therapeutic possibilities for diverse age-related conditions.
Collapse
Affiliation(s)
| | - Lucia Mendoza-Viveros
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigacíon y de Estudios Avanzados (CIE-CINVESTAV), México City, México
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México UNAM, México City, México
| | - Carolina Cid-Castro
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigacíon y de Estudios Avanzados (CIE-CINVESTAV), México City, México
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México UNAM, México City, México
| | | | | | - Ricardo Orozco-Solis
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigacíon y de Estudios Avanzados (CIE-CINVESTAV), México City, México
| |
Collapse
|
4
|
Zhang R, Liu S, Mousavi SM. Cognitive Dysfunction and Exercise: From Epigenetic to Genetic Molecular Mechanisms. Mol Neurobiol 2024; 61:6279-6299. [PMID: 38286967 DOI: 10.1007/s12035-024-03970-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024]
Abstract
Maintaining good health is crucial, and exercise plays a vital role in achieving this goal. It offers a range of positive benefits for cognitive function, regardless of age. However, as our population ages and life expectancy increases, cognitive impairment has become a prevalent issue, often coexisting with age-related neurodegenerative conditions. This can result in devastating consequences such as memory loss, difficulty speaking, and confusion, greatly hindering one's ability to lead an ordinary life. In addition, the decrease in mental capacity has a significant effect on an individual's physical and emotional well-being, greatly reducing their overall level of contentment and causing a significant financial burden for communities. While most current approaches aim to slow the decline of cognition, exercise offers a non-pharmacological, safe, and accessible solution. Its effects on cognition are intricate and involve changes in the brain's neural plasticity, mitochondrial stability, and energy metabolism. Moreover, exercise triggers the release of cytokines, playing a significant role in the body-brain connection and its impact on cognition. Additionally, exercise can influence gene expression through epigenetic mechanisms, leading to lasting improvements in brain function and behavior. Herein, we summarized various genetic and epigenetic mechanisms that can be modulated by exercise in cognitive dysfunction.
Collapse
Affiliation(s)
- Runhong Zhang
- Department of Physical Education, Luliang University, Lishi, 033000, Shanxi, China.
| | - Shangwu Liu
- Department of Physical Education, Luliang University, Lishi, 033000, Shanxi, China
| | | |
Collapse
|
5
|
Forsell P, Parrado Fernández C, Nilsson B, Sandin J, Nordvall G, Segerdahl M. Positive Allosteric Modulators of Trk Receptors for the Treatment of Alzheimer's Disease. Pharmaceuticals (Basel) 2024; 17:997. [PMID: 39204102 PMCID: PMC11357672 DOI: 10.3390/ph17080997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Neurotrophins are important regulators of neuronal and non-neuronal functions. As such, the neurotrophins and their receptors, the tropomyosin receptor kinase (Trk) family of receptor tyrosine kinases, has attracted intense research interest and their role in multiple diseases including Alzheimer's disease has been described. Attempts to administer neurotrophins to patients have been reported, but the clinical trials have so far have been hampered by side effects or a lack of clear efficacy. Thus, much of the focus during recent years has been on identifying small molecules acting as agonists or positive allosteric modulators (PAMs) of Trk receptors. Two examples of successful discovery and development of PAMs are the TrkA-PAM E2511 and the pan-Trk PAM ACD856. E2511 has been reported to have disease-modifying effects in preclinical models, whereas ACD856 demonstrates both a symptomatic and a disease-modifying effect in preclinical models. Both molecules have reached the stage of clinical development and were reported to be safe and well tolerated in clinical phase 1 studies, albeit with different pharmacokinetic profiles. These two emerging small molecules are interesting examples of possible novel symptomatic and disease-modifying treatments that could complement the existing anti-amyloid monoclonal antibodies for the treatment of Alzheimer's disease. This review aims to present the concept of positive allosteric modulators of the Trk receptors as a novel future treatment option for Alzheimer's disease and other neurodegenerative and cognitive disorders, and the current preclinical and clinical data supporting this new concept. Preclinical data indicate dual mechanisms, not only as cognitive enhancers, but also a tentative neurorestorative function.
Collapse
Affiliation(s)
- Pontus Forsell
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| | - Cristina Parrado Fernández
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| | - Boel Nilsson
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
| | - Johan Sandin
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| | - Gunnar Nordvall
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| | - Märta Segerdahl
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden; (C.P.F.); (B.N.); (J.S.); (G.N.); (M.S.)
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Alfred Nobels allé 23, 141 52 Huddinge, Sweden
| |
Collapse
|
6
|
von Bohlen Und Halbach O, Klausch M. The Neurotrophin System in the Postnatal Brain-An Introduction. BIOLOGY 2024; 13:558. [PMID: 39194496 DOI: 10.3390/biology13080558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024]
Abstract
Neurotrophins can bind to and signal through specific receptors that belong to the class of the Trk family of tyrosine protein kinase receptors. In addition, they can bind and signal through a low-affinity receptor, termed p75NTR. Neurotrophins play a crucial role in the development, maintenance, and function of the nervous system in vertebrates, but they also have important functions in the mature nervous system. In particular, they are involved in synaptic and neuronal plasticity. Thus, it is not surprisingly that they are involved in learning, memory and cognition and that disturbance in the neurotrophin system can contribute to psychiatric diseases. The neurotrophin system is sensitive to aging and changes in the expression levels correlate with age-related changes in brain functions. Several polymorphisms in genes coding for the different neurotrophins or neurotrophin receptors have been reported. Based on the importance of the neurotrophins for the central nervous system, it is not surprisingly that several of these polymorphisms are associated with psychiatric diseases. In this review, we will shed light on the functions of neurotrophins in the postnatal brain, especially in processes that are involved in synaptic and neuronal plasticity.
Collapse
Affiliation(s)
- Oliver von Bohlen Und Halbach
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich Loeffler Str. 23c, 17489 Greifswald, Germany
| | - Monique Klausch
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich Loeffler Str. 23c, 17489 Greifswald, Germany
| |
Collapse
|
7
|
Voigt MW, Schepers J, Haas J, von Bohlen Und Halbach O. Reduced Levels of Brain-Derived Neurotrophic Factor Affect Body Weight, Brain Weight and Behavior. BIOLOGY 2024; 13:159. [PMID: 38534429 DOI: 10.3390/biology13030159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/28/2024]
Abstract
Neurotrophins, which belong to the family of growth factors, not only play crucial roles during development but are also involved in many processes in the postnatal brain. One representative of neurotrophins is brain-derived neurotrophic factor (BDNF). BDNF plays a role in the regulation of body weight and neuronal plasticity and is, therefore, also involved in processes associated with learning and memory formation. Many of the studies on BDNF have been carried out using BDNF-deficient mice. Unfortunately, homozygous deletion of BDNF is lethal in the early postnatal stage, so heterozygous BDNF-deficient mice are often studied. Another possibility is the use of conditional BDNF-deficient mice in which the expression of BDNF is strongly downregulated in some brain cells, for example, in the neurons of the central nervous system, but the expression of BDNF in other cells in the brain is unchanged. To further reduce BDNF expression, we crossed heterozygous BDNF-deficient mice with mice carrying a deletion of BDNF in neurofilament L-positive neurons. These offspring are viable, and the animals with a strong reduction in BDNF in the brain show a strongly increased body weight, which is accompanied by a reduction in brain weight. In addition, these animals show behavioral abnormalities, particularly with regard to locomotion.
Collapse
Affiliation(s)
- Matthias Wilhelm Voigt
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler Str. 23c, D-17489 Greifswald, Germany
| | - Jens Schepers
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler Str. 23c, D-17489 Greifswald, Germany
| | - Jacqueline Haas
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler Str. 23c, D-17489 Greifswald, Germany
| | - Oliver von Bohlen Und Halbach
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler Str. 23c, D-17489 Greifswald, Germany
| |
Collapse
|
8
|
Goto T, Saligan LN, Li X, Xiang L, Kwiat C, Nguyen C, Crouch A, Von Ah D. Associations of brain-derived neurotrophic factor rs6265 polymorphism and cognitive function in breast cancer survivors from a cross-sectional study. Cancer Med 2024; 13:e6975. [PMID: 38379321 PMCID: PMC10839126 DOI: 10.1002/cam4.6975] [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: 07/12/2023] [Revised: 12/28/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Breast cancer survivors (BCS) often complain of cancer-related cognitive impairment (CRCI) during and even months after completing primary cancer treatments, particularly chemotherapy. The etiology of CRCI is unknown, but associations of CRCI with germline genetic polymorphisms have been reported, including Brain-Derived Neurotrophic Factor (BDNF) rs6265 polymorphism. The current study investigated the associations of specific BDNF rs6265 with CRCI. METHODS Cancer-related cognitive impairment was assessed using subjective reports of cognitive symptoms (the version 1.0, 8-item short-forms of the Patient-Reported Outcomes Measurement Information System®) and computerized objective cognitive function scores (CANTAB®). BDNF rs6265 genotypes were determined from buccal swabs. The associations of specific BDNF rs6265 with CRCI were examined by either one-way analysis of variance or the Kruskal-Wallis test followed by post hoc tests and rank-based regression analysis. RESULTS We examined 356 female BCS. The mean (SD) age was 55.6 (9.8) years old, the median (IQR) years since cancer diagnosis were 4.0 (6.0), and 331 (92.7%) were self-described as White. BCS carrying the Met/Met genotype showed poorer results on 'visual episodic memory and new learning' and 'spatial working memory and executive function.' This relationship was observed regardless of prior chemotherapy. CONCLUSION Our findings suggest that carrying the BDNF rs6265 Met/Met genotype increases the risk for CRCI in BCS. These results are foundational in nature and provide important information to identify mechanisms underpinning CRCI.
Collapse
Affiliation(s)
- Taichi Goto
- Symptoms Biology Unit, Division of Intramural ResearchNational Institute of Nursing Research, National Institutes of HealthBethesdaMarylandUSA
| | - Leorey N. Saligan
- Symptoms Biology Unit, Division of Intramural ResearchNational Institute of Nursing Research, National Institutes of HealthBethesdaMarylandUSA
| | - Xiaobai Li
- Department of BiostatisticsNational Institutes of Health Clinical CenterBethesdaMarylandUSA
| | - Lichen Xiang
- Symptoms Biology Unit, Division of Intramural ResearchNational Institute of Nursing Research, National Institutes of HealthBethesdaMarylandUSA
| | - Catherine Kwiat
- Symptoms Biology Unit, Division of Intramural ResearchNational Institute of Nursing Research, National Institutes of HealthBethesdaMarylandUSA
| | - Christopher Nguyen
- Symptoms Biology Unit, Division of Intramural ResearchNational Institute of Nursing Research, National Institutes of HealthBethesdaMarylandUSA
| | - Adele Crouch
- Indiana University School of NursingIndianapolisIndianaUSA
| | - Diane Von Ah
- The Ohio State University College of NursingColumbusOhioUSA
| |
Collapse
|
9
|
Tomás AM, Bento-Torres NVO, Jardim NYV, Moraes PM, da Costa VO, Modesto AC, Khayat AS, Bento-Torres J, Picanço-Diniz CW. Risk Polymorphisms of FNDC5, BDNF, and NTRK2 and Poor Education Interact and Aggravate Age-Related Cognitive Decline. Int J Mol Sci 2023; 24:17210. [PMID: 38139046 PMCID: PMC10743741 DOI: 10.3390/ijms242417210] [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: 10/19/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Cognitive abilities tend to decline with aging, with variation between individuals, and many studies seek to identify genetic biomarkers that more accurately anticipate risks related to pathological aging. We investigated the influence of BDNF, NTRK2, and FNDC5 single nucleotide polymorphisms (SNPs) on the cognitive performance of young and older adults with contrasting educational backgrounds. We addressed three questions: (1) Is education associated with reduced age-related cognitive decline? (2) Does the presence of SNPs explain the variation in cognitive performance observed late in life? (3) Is education differentially associated with cognition based on the presence of BDNF, NTRK2, or FNDC5 polymorphisms? We measured the cognitive functions of young and older participants, with lower and higher education, using specific and sensitive tests of the Cambridge Automated Neuropsychological Test Assessment Battery. A three-way ANOVA revealed that SNPs were associated with differential performances in executive functions, episodic memory, sustained attention, mental and motor response speed, and visual recognition memory and that higher educational levels improved the affected cognitive functions. The results revealed that distinct SNPs affect cognition late in life differentially, suggesting their utility as potential biomarkers and emphasizing the importance of cognitive stimulation that advanced education early in life provides.
Collapse
Affiliation(s)
- Alessandra Mendonça Tomás
- Neurodegeneration and Infection Research Laboratory, Institute of Biological Science, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (A.M.T.); (N.Y.V.J.); (P.M.M.); (V.O.d.C.); (J.B.-T.); (C.W.P.-D.)
- Department of Physical Education, Federal University of Pará Application School, Belém 66095-780, Brazil
| | - Natáli Valim Oliver Bento-Torres
- Neurodegeneration and Infection Research Laboratory, Institute of Biological Science, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (A.M.T.); (N.Y.V.J.); (P.M.M.); (V.O.d.C.); (J.B.-T.); (C.W.P.-D.)
- Graduate Program in Human Movement Sciences, Federal University of Pará, Belém 66095-780, Brazil
| | - Naina Yuki Vieira Jardim
- Neurodegeneration and Infection Research Laboratory, Institute of Biological Science, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (A.M.T.); (N.Y.V.J.); (P.M.M.); (V.O.d.C.); (J.B.-T.); (C.W.P.-D.)
- Graduate Program in Neuroscience and Cell Biology, Federal University of Pará, Belém 66050-160, Brazil
| | - Patrícia Martins Moraes
- Neurodegeneration and Infection Research Laboratory, Institute of Biological Science, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (A.M.T.); (N.Y.V.J.); (P.M.M.); (V.O.d.C.); (J.B.-T.); (C.W.P.-D.)
- Graduate Program in Human Movement Sciences, Federal University of Pará, Belém 66095-780, Brazil
| | - Victor Oliveira da Costa
- Neurodegeneration and Infection Research Laboratory, Institute of Biological Science, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (A.M.T.); (N.Y.V.J.); (P.M.M.); (V.O.d.C.); (J.B.-T.); (C.W.P.-D.)
- Graduate Program in Neuroscience and Cell Biology, Federal University of Pará, Belém 66050-160, Brazil
| | - Antônio Conde Modesto
- Oncology Research Center (NPO), Graduate Program in Oncology and Medical Sciences, Federal University of Pará, Belém 66073-000, Brazil; (A.C.M.); (A.S.K.)
| | - André Salim Khayat
- Oncology Research Center (NPO), Graduate Program in Oncology and Medical Sciences, Federal University of Pará, Belém 66073-000, Brazil; (A.C.M.); (A.S.K.)
| | - João Bento-Torres
- Neurodegeneration and Infection Research Laboratory, Institute of Biological Science, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (A.M.T.); (N.Y.V.J.); (P.M.M.); (V.O.d.C.); (J.B.-T.); (C.W.P.-D.)
- Graduate Program in Human Movement Sciences, Federal University of Pará, Belém 66095-780, Brazil
| | - Cristovam Wanderley Picanço-Diniz
- Neurodegeneration and Infection Research Laboratory, Institute of Biological Science, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66073-000, Brazil; (A.M.T.); (N.Y.V.J.); (P.M.M.); (V.O.d.C.); (J.B.-T.); (C.W.P.-D.)
- Graduate Program in Neuroscience and Cell Biology, Federal University of Pará, Belém 66050-160, Brazil
- Oncology Research Center (NPO), Graduate Program in Oncology and Medical Sciences, Federal University of Pará, Belém 66073-000, Brazil; (A.C.M.); (A.S.K.)
| |
Collapse
|
10
|
Parrado Fernandez C, Juric S, Backlund M, Dahlström M, Madjid N, Lidell V, Rasti A, Sandin J, Nordvall G, Forsell P. Neuroprotective and Disease-Modifying Effects of the Triazinetrione ACD856, a Positive Allosteric Modulator of Trk-Receptors for the Treatment of Cognitive Dysfunction in Alzheimer's Disease. Int J Mol Sci 2023; 24:11159. [PMID: 37446337 DOI: 10.3390/ijms241311159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
The introduction of anti-amyloid monoclonal antibodies against Alzheimer's disease (AD) is of high importance. However, even though treated patients show very little amyloid pathology, there is only a modest effect on the rate of cognitive decline. Although this effect can possibly increase over time, there is still a need for alternative treatments that will improve cognitive function in patients with AD. Therefore, the purpose of this study was to characterize the triazinetrione ACD856, a novel pan-Trk positive allosteric modulator, in multiple models to address its neuroprotective and potential disease-modifying effects. The pharmacological effect of ACD856 was tested in recombinant cell lines, primary cortical neurons, or animals. We demonstrate that ACD856 enhanced NGF-induced neurite outgrowth, increased the levels of the pre-synaptic protein SNAP25 in PC12 cells, and increased the degree of phosphorylated TrkB in SH-SY5Y cells. In primary cortical neurons, ACD856 led to increased levels of phospho-ERK1/2, showed a neuroprotective effect against amyloid-beta or energy-deprivation-induced neurotoxicity, and increased the levels of brain-derived neurotrophic factor (BDNF). Consequently, administration of ACD856 resulted in a significant increase in BDNF in the brains of 21 months old mice. Furthermore, repeated administration of ACD856 resulted in a sustained anti-depressant effect, which lasted up to seven days, suggesting effects that go beyond merely symptomatic effects. In conclusion, the results confirm ACD856 as a cognitive enhancer, but more importantly, they provide substantial in vitro and in vivo evidence of neuroprotective and long-term effects that contribute to neurotrophic support and increased neuroplasticity. Presumably, the described effects of ACD856 may improve cognition, increase resilience, and promote neurorestorative processes, thereby leading to a healthier brain in patients with AD.
Collapse
Affiliation(s)
- Cristina Parrado Fernandez
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, 171 77 Solna, Sweden
| | - Sanja Juric
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
| | - Maria Backlund
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
| | | | - Nather Madjid
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
| | | | - Azita Rasti
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
| | - Johan Sandin
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, 171 77 Solna, Sweden
| | - Gunnar Nordvall
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, 171 77 Solna, Sweden
| | - Pontus Forsell
- AlzeCure Pharma AB, Hälsovägen 7, 141 57 Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, 171 77 Solna, Sweden
| |
Collapse
|
11
|
Nicholson EL, Garry MI, Ney LJ, Hsu CMK, Zuj DV, Felmingham KL. The influence of the BDNF Val66Met genotype on emotional recognition memory in post-traumatic stress disorder. Sci Rep 2023; 13:5033. [PMID: 36977737 PMCID: PMC10050310 DOI: 10.1038/s41598-023-30787-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/01/2023] [Indexed: 03/30/2023] Open
Abstract
Dysregulated consolidation of emotional memories is a core feature of posttraumatic stress disorder (PTSD). Brain Derived Neurotrophic Factor (BDNF) influences synaptic plasticity and emotional memory consolidation. The BDNF Val66Met polymorphism has been associated with PTSD risk and memory deficits respectively, although findings have been inconsistent, potentially due to a failure to control for important confounds such as sex, ethnicity, and the timing/extent of previous trauma experiences. Furthermore, very little research has examined the impact of BDNF genotypes on emotional memory in PTSD populations. This study investigated the interaction effects of Val66Met and PTSD symptomatology in an emotional recognition memory task in 234 participants divided into healthy control (n = 85), trauma exposed (TE: n = 105) and PTSD (n = 44) groups. Key findings revealed impaired negative recognition memory in PTSD compared to control and TE groups and in participants with the Val/Met compared to the Val/Val genotype. There was a group × genotype interaction showing no Met effect in the TE group despite significant effects in PTSD and controls. Results suggest that people previously exposed to trauma who do not develop PTSD may be protected from the BDNF Met effect, however more research is needed to replicate findings and to explore the epigenetic and neural processes involved.
Collapse
Affiliation(s)
- Emma Louise Nicholson
- Melbourne School of Psychological Sciences, University of Melbourne, Redmond Barry Building, Parkville, VIC, 3010, Australia.
| | - Michael I Garry
- School of Psychological Sciences, University of Tasmania, Hobart, Australia
| | - Luke J Ney
- School of Psychological Sciences, University of Tasmania, Hobart, Australia
- Faculty of Health, School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Chia-Ming K Hsu
- School of Psychological Sciences, University of Tasmania, Hobart, Australia
| | - Daniel V Zuj
- School of Psychological Sciences, University of Tasmania, Hobart, Australia
- Experimental Psychopathology Lab, Department of Psychology, Swansea University, Swansea, UK
| | - Kim L Felmingham
- Melbourne School of Psychological Sciences, University of Melbourne, Redmond Barry Building, Parkville, VIC, 3010, Australia
| |
Collapse
|
12
|
Islas-Preciado D, Splinter TFL, Ibrahim M, Black N, Wong S, Lieblich SE, Liu-Ambrose T, Barha CK, Galea LAM. Sex and BDNF Val66Met polymorphism matter for exercise-induced increase in neurogenesis and cognition in middle-aged mice. Horm Behav 2023; 148:105297. [PMID: 36623432 DOI: 10.1016/j.yhbeh.2022.105297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/23/2022] [Accepted: 12/15/2022] [Indexed: 01/09/2023]
Abstract
Females show greater benefits of exercise on cognition in both humans and rodents, which may be related to brain-derived neurotrophic factor (BDNF). A single nucleotide polymorphism (SNP), the Val66Met polymorphism, within the human BDNF gene, causes impaired activity-dependent secretion of neuronal BDNF and impairments to some forms of memory. We evaluated whether sex and BDNF genotype (Val66Met polymorphism (Met/Met) versus wild-type (Val/Val)) influenced the ability of voluntary running to enhance cognition and hippocampal neurogenesis in mice. Middle-aged C57BL/6J (13 months) mice were randomly assigned to either a control or an aerobic training (AT) group (running disk access). Mice were trained on the visual discrimination and reversal paradigm in a touchscreen-based technology to evaluate cognitive flexibility. BDNF Met/Met mice had fewer correct responses compared to BDNF Val/Val mice on both cognitive tasks. Female BDNF Val/Val mice showed greater cognitive flexibility compared to male mice regardless of AT. Despite running less than BDNF Val/Val mice, AT improved performance in both cognitive tasks in BDNF Met/Met mice. AT increased neurogenesis in the ventral hippocampus of BDNF Val/Val mice of both sexes and increased the proportion of mature type 3 doublecortin-expressing cells in the dorsal hippocampus of female mice only. Our results indicate AT improved cognitive performance in BDNF Met/Met mice and increased hippocampal neurogenesis in BDNF Val/Val mice in middle age. Furthermore, middle-aged female mice may benefit more from AT than males in terms of neuroplasticity, an effect that was influenced by the BDNF Val66Met polymorphism.
Collapse
Affiliation(s)
- Dannia Islas-Preciado
- Department of Psychology, University of British Columbia, Canada; Dajavad Mowifaghian Centre for Brain Health, University of British Columbia, Canada; Lab de Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 14370, México
| | | | - Muna Ibrahim
- Department of Psychology, University of British Columbia, Canada
| | - Natasha Black
- Department of Psychology, University of British Columbia, Canada
| | - Sarah Wong
- Department of Psychology, University of British Columbia, Canada
| | | | - Teresa Liu-Ambrose
- Department of Physical Therapy, University of British Columbia, Canada; Dajavad Mowifaghian Centre for Brain Health, University of British Columbia, Canada
| | - Cindy K Barha
- Department of Physical Therapy, University of British Columbia, Canada; Dajavad Mowifaghian Centre for Brain Health, University of British Columbia, Canada.
| | - Liisa A M Galea
- Department of Psychology, University of British Columbia, Canada; Dajavad Mowifaghian Centre for Brain Health, University of British Columbia, Canada.
| |
Collapse
|
13
|
Nordvall G, Forsell P, Sandin J. Neurotrophin-targeted therapeutics: A gateway to cognition and more? Drug Discov Today 2022; 27:103318. [PMID: 35850433 DOI: 10.1016/j.drudis.2022.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 02/09/2023]
Abstract
Neurotrophins, such as brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), are small proteins expressed in the brain and peripheral tissues, which regulate several key aspects of neuronal function, including neurogenesis, synaptic plasticity and neuroprotection, but also programmed cell death. This broad range of effects is a result of a complex downstream signaling pathway, with differential spatial and temporal activation patterns further diversifying their physiological effects. Alterations in neurotrophin levels, or known polymorphisms in neurotrophin genes, have been linked to a variety of disorders, including depression and Alzheimer's disease (AD). Historically, their therapeutic potential in these disorders has been hampered by the lack of suitable tool molecules for clinical studies. However, recent advancements have led to the development of new therapeutic candidates, which are now in clinical testing.
Collapse
|
14
|
Dresang HC, Harvey DY, Xie SX, Shah-Basak PP, DeLoretta L, Wurzman R, Parchure SY, Sacchetti D, Faseyitan O, Lohoff FW, Hamilton RH. Genetic and Neurophysiological Biomarkers of Neuroplasticity Inform Post-Stroke Language Recovery. Neurorehabil Neural Repair 2022; 36:371-380. [PMID: 35428413 PMCID: PMC9133188 DOI: 10.1177/15459683221096391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND There is high variability in post-stroke aphasia severity and predicting recovery remains imprecise. Standard prognostics do not include neurophysiological indicators or genetic biomarkers of neuroplasticity, which may be critical sources of variability. OBJECTIVE To evaluate whether a common polymorphism (Val66Met) in the gene for brain-derived neurotrophic factor (BDNF) contributes to variability in post-stroke aphasia, and to assess whether BDNF polymorphism interacts with neurophysiological indicators of neuroplasticity (cortical excitability and stimulation-induced neuroplasticity) to improve estimates of aphasia severity. METHODS Saliva samples and motor-evoked potentials (MEPs) were collected from participants with chronic aphasia subsequent to left-hemisphere stroke. MEPs were collected prior to continuous theta burst stimulation (cTBS; index for cortical excitability) and 10 minutes following cTBS (index for stimulation-induced neuroplasticity) to the right primary motor cortex. Analyses assessed the extent to which BDNF polymorphism interacted with cortical excitability and stimulation-induced neuroplasticity to predict aphasia severity beyond established predictors. RESULTS Val66Val carriers showed less aphasia severity than Val66Met carriers, after controlling for lesion volume and time post-stroke. Furthermore, Val66Val carriers showed expected effects of age on aphasia severity, and positive associations between severity and both cortical excitability and stimulation-induced neuroplasticity. In contrast, Val66Met carriers showed weaker effects of age and negative associations between cortical excitability, stimulation-induced neuroplasticity and aphasia severity. CONCLUSIONS Neurophysiological indicators and genetic biomarkers of neuroplasticity improved aphasia severity predictions. Furthermore, BDNF polymorphism interacted with cortical excitability and stimulation-induced neuroplasticity to improve predictions. These findings provide novel insights into mechanisms of variability in stroke recovery and may improve aphasia prognostics.
Collapse
Affiliation(s)
- Haley C. Dresang
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104,Moss Rehabilitation Research Institute, Einstein Medical Center, 50 Township Line Road, Philadelphia, PA 19027,Corresponding author:, Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104
| | - Denise Y. Harvey
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104
| | - Sharon Xiangwen Xie
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Perelman School of Medicine, 607 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104
| | - Priyanka P. Shah-Basak
- Medical College of Wisconsin, Department of Neurology, 8701 Watertown Plank Road Milwaukee, WI 53226
| | - Laura DeLoretta
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104
| | - Rachel Wurzman
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104
| | - Shreya Y. Parchure
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104
| | - Daniela Sacchetti
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104
| | - Olufunsho Faseyitan
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104
| | - Falk W. Lohoff
- National Institute for Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), 10 Center Drive (10CRC/2-2352), Bethesda, MD 20892
| | - Roy H. Hamilton
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, 3710 Hamilton Walk, Philadelphia, PA 19104
| |
Collapse
|
15
|
Huang X, Xie Z, Wang C, Wang S. Elevated Peripheral Brain-Derived Neurotrophic Factor Level Associated With Decreasing Insulin Secretion May Forecast Memory Dysfunction in Patients With Long-Term Type 2 Diabetes. Front Physiol 2022; 12:686838. [PMID: 35111074 PMCID: PMC8801615 DOI: 10.3389/fphys.2021.686838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Background With the progressive course of diabetes and the decline in islet function, the cognitive dysfunction of patients aggravated. Objective We aimed to investigate the roles of brain-derived neurotrophic factor (BDNF) and the Val66Met polymorphism in mild cognitive impairment (MCI) in patients with type 2 diabetes mellitus (T2DM). Methods A total of 169 Chinese patients with T2DM were involved and divided into long-term (diabetes duration >10 years) and short-term (diabetes duration ≤10 years) diabetes, and in each group, the patients were separated as MCI and the control. Demographic characteristics, clinical variables, and cognitive performances were assessed. The plasma BDNF level was measured via enzyme-linked immunosorbent assay. The Val66Met polymorphisms were analyzed. Results Long-term T2DM have lower 2 h postprandial C-peptide (p < 0.05). The BDNF level was slightly higher in patients with MCI than in the controls in each duration group without statistical significance. The relationship of BDNF to Montreal Cognitive Assessment was not proven either. However, in the long-term diabetes group, BDNF concentration remained as an independent factor of logical memory test (β = −0.27; p < 0.05), and they were negatively correlated (r = −0.267; p = 0.022); BDNF was also negatively correlated with fasting C-peptide (r = −0.260; p = 0.022), 2 h postprandial C-peptide (r = −0.251; p = 0.028), and homeostasis model assessment of insulin resistance (r = −0.312; p = 0.006). In genotypic groups, BDNF Val/Val performed better in logical memory test than Met/Met and Val/Met. Conclusion Elevated peripheral BDNF level associated with declined islet function, when combined with its Val66Met polymorphism, may forecast memory dysfunction in patients with long-term T2DM.
Collapse
Affiliation(s)
- Xi Huang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Zuolin Xie
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Chenchen Wang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Shaohua Wang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
- Nanjing Medical University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- *Correspondence: Shaohua Wang,
| |
Collapse
|
16
|
BDNF Val66Met gene polymorphism modulates brain activity following rTMS-induced memory impairment. Sci Rep 2022; 12:176. [PMID: 34997117 PMCID: PMC8741781 DOI: 10.1038/s41598-021-04175-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/16/2021] [Indexed: 01/19/2023] Open
Abstract
The BDNF Val66Met gene polymorphism is a relevant factor explaining inter-individual differences to TMS responses in studies of the motor system. However, whether this variant also contributes to TMS-induced memory effects, as well as their underlying brain mechanisms, remains unexplored. In this investigation, we applied rTMS during encoding of a visual memory task either over the left frontal cortex (LFC; experimental condition) or the cranial vertex (control condition). Subsequently, individuals underwent a recognition memory phase during a functional MRI acquisition. We included 43 young volunteers and classified them as 19 Met allele carriers and 24 as Val/Val individuals. The results revealed that rTMS delivered over LFC compared to vertex stimulation resulted in reduced memory performance only amongst Val/Val allele carriers. This genetic group also exhibited greater fMRI brain activity during memory recognition, mainly over frontal regions, which was positively associated with cognitive performance. We concluded that BDNF Val66Met gene polymorphism, known to exert a significant effect on neuroplasticity, modulates the impact of rTMS both at the cognitive as well as at the associated brain networks expression levels. This data provides new insights on the brain mechanisms explaining cognitive inter-individual differences to TMS, and may inform future, more individually-tailored rTMS interventions.
Collapse
|
17
|
West GL, Konishi K, MacDonald K, Ni A, Joober R, Bohbot VD. The BDNF val66met polymorphism is associated with decreased use of landmarks and decreased fMRI activity in the hippocampus during virtual navigation. Eur J Neurosci 2021; 54:6406-6421. [PMID: 34467592 DOI: 10.1111/ejn.15431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/01/2022]
Abstract
People can navigate in a new environment using multiple strategies dependent on different memory systems. A series of studies have dissociated between hippocampus-dependent 'spatial' navigation and habit-based 'response' learning mediated by the caudate nucleus. The val66met polymorphism of the brain-derived neurotrophic factor (BDNF) gene leads to decreased secretion of BDNF in the brain, including the hippocampus. Here, we aim to investigate the role of the BDNF val66met polymorphism on virtual navigation behaviour and brain activity in healthy older adults. A total of 139 healthy older adult participants (mean age = 65.8 ± 4.4 years) were tested in this study. Blood samples were collected, and BDNF val66met genotyping was performed. Participants were divided into two genotype groups: val homozygotes and met carriers. Participants were tested on virtual dual-solution navigation tasks in which they could use either a hippocampus-dependent spatial strategy or a caudate nucleus-dependent response strategy to solve the task. A subset of the participants (n = 66) were then scanned in a 3T functional magnetic resonance imaging (fMRI) scanner while engaging in another dual-solution navigation task. BDNF val/val individuals and met carriers did not differ in learning performance. However, the two BDNF groups differed in learning strategy. BDNF val/val individuals relied more on landmarks to remember target locations (i.e., increased use of flexible spatial learning), while met carriers relied more on sequences and patterns to remember target locations (i.e., increased use of inflexible response learning). Additionally, BDNF val/val individuals had more fMRI activity in the hippocampus compared with BDNF met carriers during performance on the navigation task. This is the first study to show in older adults that BDNF met carriers use alternate learning strategies from val/val individuals and to identify differential brain activation of this behavioural difference between the two groups.
Collapse
Affiliation(s)
- Greg L West
- Department of Psychology, University of Montreal, Montréal, Quebec, Canada
| | - Kyoko Konishi
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Verdun, Quebec, Canada
| | - Kathleen MacDonald
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Verdun, Quebec, Canada
| | - Anjie Ni
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Verdun, Quebec, Canada
| | - Ridha Joober
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Verdun, Quebec, Canada
| | - Veronique D Bohbot
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Verdun, Quebec, Canada
| |
Collapse
|
18
|
Baciu M, Banjac S, Roger E, Haldin C, Perrone-Bertolotti M, Lœvenbruck H, Démonet JF. Strategies and cognitive reserve to preserve lexical production in aging. GeroScience 2021; 43:1725-1765. [PMID: 33970414 PMCID: PMC8492841 DOI: 10.1007/s11357-021-00367-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/09/2021] [Indexed: 10/28/2022] Open
Abstract
In the absence of any neuropsychiatric condition, older adults may show declining performance in several cognitive processes and among them, in retrieving and producing words, reflected in slower responses and even reduced accuracy compared to younger adults. To overcome this difficulty, healthy older adults implement compensatory strategies, which are the focus of this paper. We provide a review of mainstream findings on deficient mechanisms and possible neurocognitive strategies used by older adults to overcome the deleterious effects of age on lexical production. Moreover, we present findings on genetic and lifestyle factors that might either be protective or risk factors of cognitive impairment in advanced age. We propose that "aging-modulating factors" (AMF) can be modified, offering prevention opportunities against aging effects. Based on our review and this proposition, we introduce an integrative neurocognitive model of mechanisms and compensatory strategies for lexical production in older adults (entitled Lexical Access and Retrieval in Aging, LARA). The main hypothesis defended in LARA is that cognitive aging evolves heterogeneously and involves complementary domain-general and domain-specific mechanisms, with substantial inter-individual variability, reflected at behavioral, cognitive, and brain levels. Furthermore, we argue that the ability to compensate for the effect of cognitive aging depends on the amount of reserve specific to each individual which is, in turn, modulated by the AMF. Our conclusion is that a variety of mechanisms and compensatory strategies coexist in the same individual to oppose the effect of age. The role of reserve is pivotal for a successful coping with age-related changes and future research should continue to explore the modulating role of AMF.
Collapse
Affiliation(s)
- Monica Baciu
- Univ. Grenoble Alpes, CNRS LPNC UMR 5105, 38000, Grenoble, France.
| | - Sonja Banjac
- Univ. Grenoble Alpes, CNRS LPNC UMR 5105, 38000, Grenoble, France
| | - Elise Roger
- Univ. Grenoble Alpes, CNRS LPNC UMR 5105, 38000, Grenoble, France
| | - Célise Haldin
- Univ. Grenoble Alpes, CNRS LPNC UMR 5105, 38000, Grenoble, France
| | | | | | | |
Collapse
|
19
|
Cechova K, Andel R, Angelucci F, Chmatalova Z, Markova H, Laczó J, Vyhnalek M, Matoska V, Kaplan V, Nedelska Z, Ward DD, Hort J. Impact of APOE and BDNF Val66Met Gene Polymorphisms on Cognitive Functions in Patients with Amnestic Mild Cognitive Impairment. J Alzheimers Dis 2021; 73:247-257. [PMID: 31771052 DOI: 10.3233/jad-190464] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Apolipoprotein (APOE) ɛ4 is a well-known risk factor for late-onset Alzheimer's disease (AD), but other AD-related gene polymorphisms might also be important, such as the polymorphism within the brain-derived neurotrophic factor (BDNF) gene. Carriage of BDNF Val66Met has been associated with faster cognitive decline and greater hippocampal atrophy in cognitively normal elderly. Thus, we examined the effects of the concurrent presence of APOE and BDNF polymorphisms on cognitive functions and brain morphometry in amnestic mild cognitive impairment (aMCI) patients. 107 aMCI patients (mean age = 72.2) were recruited from the Czech Brain Aging Study and, based on APOE and BDNF genes polymorphisms, were divided into four groups: ɛ4-BDNFVal/Val (n = 37), ɛ4-BDNFMet (n = 19), ɛ4+BDNFVal/Val (n = 35), and ɛ4+BDNFMet (n = 16). All patients underwent clinical examination, magnetic resonance imaging, and complex neuropsychological battery. The combination of APOEɛ4+ and BDNF Met was associated with significantly worse memory performance in immediate and delayed recall compared to other polymorphism groups. We did not observe increased atrophy in areas related to memory function in the ɛ4+BDNFMet group. Our findings suggest that carriage of ɛ4+BDNFMet is associated with more pronounced memory dysfunction, a typical feature of early AD, but not with structural brain changes in aMCI patients. These findings suggest that in APOEɛ4/BDNF Met carriers, synaptic dysfunction affecting memory may precede pronounced structural changes.
Collapse
Affiliation(s)
- Katerina Cechova
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Ross Andel
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,School of Aging Studies, University of South Florida, Tampa, FL, USA
| | - Francesco Angelucci
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Zuzana Chmatalova
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hana Markova
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jan Laczó
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Martin Vyhnalek
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Vaclav Matoska
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Vojtech Kaplan
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Zuzana Nedelska
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - David D Ward
- Department of Medicine, Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,Centre for Health Care of the Elderly, QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Jakub Hort
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
20
|
Braun RG, Kittner SJ, Ryan KA, Cole JW. Effects of the BDNF Val66Met polymorphism on functional status and disability in young stroke patients. PLoS One 2020; 15:e0237033. [PMID: 33306691 PMCID: PMC7732081 DOI: 10.1371/journal.pone.0237033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/17/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE The preponderance of evidence from recent studies in human subjects supports a negative effect of the BDNF Val66Met polymorphism on motor outcomes and motor recovery. However prior studies have generally reported the effect of the Met allele in older stroke patients, while potential effects in younger stroke patients have remained essentially unexamined. The lack of research in younger patients is significant since aging effects on CNS repair and functional recovery after stroke are known to interact with the effects of genetic polymorphisms. Here we present a study of first-ever ischemic stroke patients aged 15-49 years that examines the effect of Met carrier status on functional disability. METHODS 829 patients with a first ischemic stroke (Average age = 41.4 years, SD = 6.9) were recruited from the Baltimore-Washington region. Genotyping was performed at the Johns Hopkins University Center for Inherited Disease Research (CIDR). Data cleaning and harmonization were done at the GEI-funded GENEVA Coordinating Center at the University of Washington. Our sample contained 165 Met carriers and 664 non-Met carriers. Modified Rankin scores as recorded at discharge were obtained from the hospital records by study personnel blinded to genotype, and binarized into "Good" versus "Poor" outcomes (mRS 0-2 vs. 3+), with mRS scores 3+ reflecting a degree of disability that causes loss of independence. RESULTS Our analysis showed that the Met allele conveyed a proportionally greater risk for poor outcomes and disability-related loss of independence with mRS scores 3+ (adjusted OR 1.73, 95% CI 1.13-2.64, p = 0.01). CONCLUSIONS The BDNF Val66Met polymorphism was negatively associated with functional outcomes at discharge in our sample of 829 young stroke patients. This finding stands in contrast to what would be predicted under the tenets of the resource modulation hypothesis (i.e. that younger patients would be spared from the negative effect of the Met allele on recovery since it is posited to arise as a manifestation of age-related decline in physiologic resources).
Collapse
Affiliation(s)
- Robynne G. Braun
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Steven J. Kittner
- University of Maryland School of Medicine, Baltimore, MD, United States of America
- Veterans Affairs Maryland Health Care System, Baltimore, MD, United States of America
| | - Kathleen A. Ryan
- Veterans Affairs Maryland Health Care System, Baltimore, MD, United States of America
| | - John W. Cole
- University of Maryland School of Medicine, Baltimore, MD, United States of America
- Veterans Affairs Maryland Health Care System, Baltimore, MD, United States of America
| |
Collapse
|
21
|
Boroujeni NB, Ashkezari MD, Seifati SM. The rs6265 polymorphism might not affect the secretion of BDNF protein directedly. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
22
|
Foster CM, Kennedy KM, Daugherty AM, Rodrigue KM. Contribution of iron and Aβ to age differences in entorhinal and hippocampal subfield volume. Neurology 2020; 95:e2586-e2594. [PMID: 32938781 PMCID: PMC7682827 DOI: 10.1212/wnl.0000000000010868] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 06/26/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To test the hypothesis that the combination of elevated global β-AMYLOID (Aβ) burden and greater striatal iron content would be associated with smaller entorhinal cortex (ERC) volume, but not hippocampal subfield volumes, we measured volume and iron content using high-resolution MRI and Aβ using PET imaging in a cross-sectional sample of 70 cognitively normal older adults. METHODS Participants were scanned with florbetapir 18F PET to obtain Aβ standardized uptake value ratios. Susceptibility-weighted MRI was collected and processed to yield R2* images, and striatal regions of interest (ROIs) were manually placed to obtain a measure of striatal iron burden. Ultra-high resolution T2/PD-weighted MRIs were segmented to measure medial temporal lobe (MTL) volumes. Analyses were conducted using mixed-effects models with MTL ROI as a within-participant factor; age, iron content, and Aβ as between-participant factors; and MTL volumes (ERC and 3 hippocampal subfield regions) as the dependent variable. RESULTS The model indicated a significant 4-way interaction among age, iron, Aβ, and MTL region. Post hoc analyses indicated that the 3-way interaction among age, Aβ, and iron content was selective to the ERC (β = -3.34, standard error = 1.33, 95% confidence interval -5.95 to -0.72), whereas a significant negative association between age and ERC volume was present only in individuals with both elevated iron content and Aβ. CONCLUSIONS These findings highlight the importance of studying Aβ in the context of other, potentially synergistic age-related brain factors such as iron accumulation and the potential role for iron as an important contributor to the earliest, preclinical stages of pathologic aging.
Collapse
Affiliation(s)
- Chris M Foster
- From the School of Behavioral and Brain Sciences (C.M.F., K.M.K., K.M.R.), Center for Vital Longevity, University of Texas at Dallas; and Department of Psychology (A.M.D.) and Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI
| | - Kristen M Kennedy
- From the School of Behavioral and Brain Sciences (C.M.F., K.M.K., K.M.R.), Center for Vital Longevity, University of Texas at Dallas; and Department of Psychology (A.M.D.) and Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI
| | - Ana M Daugherty
- From the School of Behavioral and Brain Sciences (C.M.F., K.M.K., K.M.R.), Center for Vital Longevity, University of Texas at Dallas; and Department of Psychology (A.M.D.) and Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI
| | - Karen M Rodrigue
- From the School of Behavioral and Brain Sciences (C.M.F., K.M.K., K.M.R.), Center for Vital Longevity, University of Texas at Dallas; and Department of Psychology (A.M.D.) and Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI.
| |
Collapse
|
23
|
Sanders CL, Rattinger GB, Deberard MS, Hammond AG, Wengreen H, Kauwe JSK, Buhusi M, Tschanz JT. Interaction Between Physical Activity and Genes Related to Neurotrophin Signaling in Late-Life Cognitive Performance: The Cache County Study. J Gerontol A Biol Sci Med Sci 2020; 75:1633-1642. [PMID: 31504225 PMCID: PMC7494026 DOI: 10.1093/gerona/glz200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Indexed: 01/23/2023] Open
Abstract
Research indicates that lifestyle and genetic factors influence the course of cognitive impairment in aging, but their interactions have not been well-examined. This study examined the relationship between physical activity and genotypes related to brain-derived neurotrophic factor (BDNF) in predicting cognitive performance in a sample of older adults with up to 12 years of follow-up. Physical activity levels (sedentary, light, and moderate/vigorous) were determined for the sample of 3,591 participants (57% female) without dementia. The genotypes examined included BDNF gene single nucleotide polymorphisms (SNPs) (rs6265 and rs56164415) and receptor gene SNPs (NTRK2 rs2289656 and NGFR rs2072446). Cognition was assessed triennially using the Modified Mini-Mental State Exam. Unadjusted linear mixed models indicated that sedentary (β = -5.05) and light (β = -2.41) groups performed worse than moderate-vigorous (p < .001). Addition of interaction effects showed significant differences in rate of decline between activity levels, particularly among males (p = .006). A three-way interaction with sex, NGFR SNP rs2072446, and physical activity suggested that the C/C allele was associated with better cognitive performance among males engaging in light activity only (p = .004). Physical activity and sex, but not BDNF-related SNPs, predicted rate of cognitive decline in older adults, while NGFR rs2072446 may modify main effects.
Collapse
Affiliation(s)
| | - Gail B Rattinger
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York
| | | | | | - Heidi Wengreen
- Dietetics and Food Sciences Department, Utah State University, Logan
| | - John S K Kauwe
- Biology Department, Brigham Young University, Provo, Utah
| | - Mona Buhusi
- Department of Psychology, Utah State University, Logan
| | | |
Collapse
|
24
|
Webb CE, Foster CM, Horn MM, Kennedy KM, Rodrigue KM. Beta-amyloid burden predicts poorer mnemonic discrimination in cognitively normal older adults. Neuroimage 2020; 221:117199. [PMID: 32736001 PMCID: PMC7813158 DOI: 10.1016/j.neuroimage.2020.117199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/29/2020] [Accepted: 07/22/2020] [Indexed: 11/18/2022] Open
Abstract
One of the earliest indicators of Alzheimer's disease pathology is the presence of beta-amyloid (Αβ) protein deposition. Significant amyloid deposition is evident even in older adults who exhibit little or no overt cognitive or memory impairment. Hippocampal-based processes that help distinguish between highly similar memory representations may be the most susceptible to early disease pathology. Amyloid associations with memory have been difficult to establish, possibly because typical memory assessments do not tax hippocampal operations sufficiently. Thus, the present study utilized a spatial mnemonic discrimination task designed to tax hippocampal pattern separation/completion processes in a sample of cognitively normal middle-aged and older adults (53-98 years old) who underwent PET 18F-Florbetapir Αβ scanning. The degree of interference between studied and new information varied, allowing for an examination of mnemonic discrimination as a function of mnemonic similarity. Results indicated that greater beta-amyloid burden was associated with poorer discrimination across decreasing levels of interference, suggesting that even subtle elevation of beta-amyloid in cognitively normal adults is associated with impoverished performance on a hippocampally demanding memory task. The present study demonstrates that degree of amyloid burden negatively impacts the ability of aging adults to accurately distinguish old from increasingly distinct new information, providing novel insight into the cognitive expression of beta-amyloid neuropathology.
Collapse
Affiliation(s)
- Christina E Webb
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 1600 Viceroy Dr., Suite 800, Dallas, TX 75235, USA
| | - Chris M Foster
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 1600 Viceroy Dr., Suite 800, Dallas, TX 75235, USA
| | - Marci M Horn
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 1600 Viceroy Dr., Suite 800, Dallas, TX 75235, USA
| | - Kristen M Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 1600 Viceroy Dr., Suite 800, Dallas, TX 75235, USA
| | - Karen M Rodrigue
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 1600 Viceroy Dr., Suite 800, Dallas, TX 75235, USA.
| |
Collapse
|
25
|
The impact of BDNF Val66Met on cognitive skills in veterans with posttraumatic stress disorder. Neurosci Lett 2020; 735:135235. [PMID: 32629065 DOI: 10.1016/j.neulet.2020.135235] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/14/2020] [Accepted: 07/03/2020] [Indexed: 12/15/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a trauma-induced disorder characterized with impaired cognitive function. BDNF modulates cognition and is involved in neuroprotection and neurocognitive processing. The BDNF Val66Met polymorphism was found to influence cognitive functions. In PTSD, carriers of the BDNF GG genotype had better spatial processing of navigation performance, and lower hyperarousal and startle reaction than A allele carriers. The hypothesis was that veterans with PTSD, carriers of the BDNF Val66Met A allele, will show reduced cognitive skills. The study included 315 male Caucasian combat veterans, with (N = 199) or without (N = 116) current and chronic PTSD. Cognition was assessed using the Rey-Osterrieth Complex Figure (ROCF) test that determines visual-spatial perception and short and long-term visual memory function. The results revealed that cognitive decline measured with ROCF test was associated with PTSD. Presence of the BDNF Val66Met GG genotype in veterans with PTSD, but not in veterans without PTSD, showed protective association with visual short-term memory and visual object manipulation after few seconds (executive function), assessed with the ROCF immediate recall test, compared to the A carriers with PTSD. In conclusion, this was the first study to confirm the association between BDNF Val66Met and memory and attention performed with ROCF in male veterans with PTSD. The results corroborated that the BDNF Val66Met A allele, compared to GG genotype, is associated with poorer short-term visual memory and attention linked with executive functions, in veterans with PTSD.
Collapse
|
26
|
Bessi V, Mazzeo S, Bagnoli S, Padiglioni S, Carraro M, Piaceri I, Bracco L, Sorbi S, Nacmias B. The implication of BDNF Val66Met polymorphism in progression from subjective cognitive decline to mild cognitive impairment and Alzheimer's disease: a 9-year follow-up study. Eur Arch Psychiatry Clin Neurosci 2020; 270:471-482. [PMID: 31560105 DOI: 10.1007/s00406-019-01069-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
Abstract
Brain-derived natriuretic factor (BDNF) Val66Met polymorphism has been frequently reported to be associated with Alzheimer's disease (AD) with contrasting results. Numerous studies showed that Met allele increased the risk of AD only in women, while other studies have found worse cognitive performance in Val/Val carriers. We aimed to inquire the effects of Val66Met polymorphism on the progression from subjective cognitive decline (SCD) to mild cognitive impairment (MCI) and from MCI to AD and to ascertain if this effect is modulated by demographic and cognitive variables. For this purpose, we followed up 74 subjects (48 SCD, 26 MCI) for a mean time of 9 years. All participants underwent extensive neuropsychological assessment, cognitive reserve estimation, BDNF and apolipoprotein E (ApoE) genotype analysis at baseline. Personality traits and leisure activities were assessed in a subgroup. Each patient underwent clinical-neuropsychological follow-up, during which 18 out of 48 SCD subjects progressed to MCI and 14 out of 26 MCI subjects progressed to AD. We found that Val66Met increased the risk of progression from SCD to MCI and from MCI to AD only in women. Nevertheless, Val/Val carriers who progressed from SCD to MCI had a shorter conversion time compared to Met carriers. We concluded that Val66Met polymorphism might play different roles depending on sex and stage of the disease.
Collapse
Affiliation(s)
- Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy.
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Sonia Padiglioni
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Marco Carraro
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Laura Bracco
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| |
Collapse
|
27
|
The BDNF Val66Met Polymorphism Modulates Resilience of Neurological Functioning to Brain Ageing and Dementia: A Narrative Review. Brain Sci 2020; 10:brainsci10040195. [PMID: 32218234 PMCID: PMC7226504 DOI: 10.3390/brainsci10040195] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023] Open
Abstract
Brain-derived neurotropic factor (BDNF) is an abundant and multi-function neurotrophin in the brain. It is released following neuronal activity and is believed to be particularly important in strengthening neural networks. A common variation in the BDNF gene, a valine to methionine substitution at codon 66 (Val66Met), has been linked to differential expression of BDNF associated with experience-dependent plasticity. The Met allele has been associated with reduced production of BDNF following neuronal stimulation, which suggests a potential role of this variation with respect to how the nervous system may respond to challenges, such as brain ageing and related neurodegenerative conditions (e.g., dementia and Alzheimer’s disease). The current review examines the potential of the BDNF Val66Met variation to modulate an individual’s susceptibility and trajectory through cognitive changes associated with ageing and dementia. On balance, research to date indicates that the BDNF Met allele at this codon is potentially associated with a detrimental influence on the level of cognitive functioning in older adults and may also impart increased risk of progression to dementia. Furthermore, recent studies also show that this genetic variation may modulate an individual’s response to interventions targeted at building cognitive resilience to conditions that cause dementia.
Collapse
|
28
|
Han Z, Qi L, Xu Q, Xu M, Cai L, Wong J, Hu X, Luo X, Wang J, Zhang Y, Li Y, Wang QM. BDNF Met allele Is Associated With Lower Cognitive Function in Poststroke Rehabilitation. Neurorehabil Neural Repair 2020; 34:247-259. [PMID: 32009534 DOI: 10.1177/1545968320902127] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background and purpose. The identification of a genetic role for cognitive outcome could influence the design of individualized treatment in poststroke rehabilitation. The aim of this study is to determine whether brain-derived neurotrophic factor ( BDNF) Val66Met polymorphism is independently associated with poststroke functional outcome. Methods. A total of 775 stroke patients with genomic data were identified from the Partners HealthCare Biobank, which contains a large number of genotypes from Biobank’s consented patients. Of 775 stroke patients who met the inclusion/exclusion criteria, 86 were enrolled. Functional outcomes were assessed using the Functional Independence Measure scores at the time of admission and discharge. Logistic and linear regression models adjusted for covariate variables, including age, sex, and medical conditions, were used to evaluate the association between BDNF Val66Met and functional outcome. Results. We detected a significant correlation between Met alleles and lower cognitive function at discharge in both ischemic and hemorrhagic stroke patients. Genotyping findings confirmed that BDNF Met allele frequency was higher in contrast to Val/Val allele frequency in lower cognitive functional recovery. Furthermore, after adjusting for covariate variables, BDNF Met alleles were found to be associated with lower cognitive outcome [ P = .003; odds ratio (OR) = 5.95 (1.81-19.52)] and recovery [ P = .006; OR = 3.16 (1.4-7.15)], especially with lower problem solving, expression, and social recovery in all stroke patients. Conclusions. Met allele carriers exhibited impaired poststroke cognitive function. The BDNF genotype may be a useful predictor of cognitive function in inpatient poststroke rehabilitation.
Collapse
Affiliation(s)
- Zhenxiang Han
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA
- Seventh People’s Hospital of Shanghai University of TCM, Shanghai, PR China
| | - Lili Qi
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of TCM, Shanghai, PR China
| | | | - Mingzhu Xu
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA
- Southern Medical University, Shenzhen, PR China
| | - Lei Cai
- Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - John Wong
- MGH Institute of Health Professions, Boston, MA, USA
| | - Xinjia Hu
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA
- Shenzhen People’s Hospital, Clinical Medical College of Jinan University, Shenzhen, Guangdong Province, PR China
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, Guangdong, PR China
- Dapeng New District Nan’ao People’s Hospital, Shenzhen, Guangdong, PR China
| | - Jiening Wang
- Seventh People’s Hospital of Shanghai University of TCM, Shanghai, PR China
| | - Yuling Zhang
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA
- University of Shanghai for Science and Technology, Shanghai, PR China
| | - Yapeng Li
- Fudan University, Shanghai, PR China
| | - Qing Mei Wang
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
29
|
Validation of a priori candidate Alzheimer's disease SNPs with brain amyloid-beta deposition. Sci Rep 2019; 9:17069. [PMID: 31745181 PMCID: PMC6863876 DOI: 10.1038/s41598-019-53604-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
The accumulation of brain amyloid β (Aβ) is one of the main pathological hallmarks of Alzheimer’s disease (AD). However, the role of brain amyloid deposition in the development of AD and the genetic variants associated with this process remain unclear. In this study, we sought to identify associations between Aβ deposition and an a priori evidence based set of 1610 genetic markers, genotyped from 505 unrelated individuals (258 Aβ+ and 247 Aβ−) enrolled in the Australian Imaging, Biomarker & Lifestyle (AIBL) study. We found statistically significant associations for 6 markers located within intronic regions of 6 genes, including AC103796.1-BDNF, PPP3R1, NGFR, KL, ABCA7 & CALHM1. Although functional studies are required to elucidate the role of these genes in the accumulation of Aβ and their potential implication in AD pathophysiology, our findings are consistent with results obtained in previous GWAS efforts.
Collapse
|
30
|
Brivio P, Paladini MS, Racagni G, Riva MA, Calabrese F, Molteni R. From Healthy Aging to Frailty: In Search of the Underlying Mechanisms. Curr Med Chem 2019; 26:3685-3701. [PMID: 31333079 DOI: 10.2174/0929867326666190717152739] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/14/2018] [Accepted: 03/08/2019] [Indexed: 11/22/2022]
Abstract
Population aging is accelerating rapidly worldwide, from 461 million people older than 65 years in 2004 to an estimated 2 billion people by 2050, leading to critical implications for the planning and delivery of health and social care. The most problematic expression of population aging is the clinical condition of frailty, which is a state of increased vulnerability that develops as a consequence of the accumulation of microscopic damages in many physiological systems that lead to a striking and disproportionate change in health state, even after an apparently small insult. Since little is known about the biology of frailty, an important perspective to understand this phenomenon is to establish how the alterations that physiologically occur during a condition of healthy aging may instead promote cumulative decline with subsequent depletion of homoeostatic reserve and increase the vulnerability also after minor stressor events. In this context, the present review aims to provide a description of the molecular mechanisms that, by having a critical impact on behavior and neuronal function in aging, might be relevant for the development of frailty. Moreover, since these biological systems are also involved in the coping strategies set in motion to respond to environmental challenges, we propose a role for lifestyle stress as an important player to drive frailty in aging.
Collapse
Affiliation(s)
- Paola Brivio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maria Serena Paladini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giorgio Racagni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.,Associazione di Psicofarmacologia, Milan, Italy
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
31
|
Miranda M, Morici JF, Zanoni MB, Bekinschtein P. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front Cell Neurosci 2019; 13:363. [PMID: 31440144 PMCID: PMC6692714 DOI: 10.3389/fncel.2019.00363] [Citation(s) in RCA: 845] [Impact Index Per Article: 140.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
Brain Derived Neurotrophic Factor (BDNF) is a key molecule involved in plastic changes related to learning and memory. The expression of BDNF is highly regulated, and can lead to great variability in BDNF levels in healthy subjects. Changes in BDNF expression are associated with both normal and pathological aging and also psychiatric disease, in particular in structures important for memory processes such as the hippocampus and parahippocampal areas. Some interventions like exercise or antidepressant administration enhance the expression of BDNF in normal and pathological conditions. In this review, we will describe studies from rodents and humans to bring together research on how BDNF expression is regulated, how this expression changes in the pathological brain and also exciting work on how interventions known to enhance this neurotrophin could have clinical relevance. We propose that, although BDNF may not be a valid biomarker for neurodegenerative/neuropsychiatric diseases because of its disregulation common to many pathological conditions, it could be thought of as a marker that specifically relates to the occurrence and/or progression of the mnemonic symptoms that are common to many pathological conditions.
Collapse
Affiliation(s)
- Magdalena Miranda
- Laboratory of Memory Research and Molecular Cognition, Institute for Cognitive and Translational Neuroscience, Instituto de Neurología Cognitiva, CONICET, Universidad Favaloro, Buenos Aires, Argentina
| | - Juan Facundo Morici
- Laboratory of Memory Research and Molecular Cognition, Institute for Cognitive and Translational Neuroscience, Instituto de Neurología Cognitiva, CONICET, Universidad Favaloro, Buenos Aires, Argentina
| | - María Belén Zanoni
- Laboratory of Memory Research and Molecular Cognition, Institute for Cognitive and Translational Neuroscience, Instituto de Neurología Cognitiva, CONICET, Universidad Favaloro, Buenos Aires, Argentina
| | - Pedro Bekinschtein
- Laboratory of Memory Research and Molecular Cognition, Institute for Cognitive and Translational Neuroscience, Instituto de Neurología Cognitiva, CONICET, Universidad Favaloro, Buenos Aires, Argentina
| |
Collapse
|
32
|
Korthauer LE, Awe E, Frahmand M, Driscoll I. Genetic Risk for Age-Related Cognitive Impairment Does Not Predict Cognitive Performance in Middle Age. J Alzheimers Dis 2019; 64:459-471. [PMID: 29865048 DOI: 10.3233/jad-171043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is characterized by memory loss and executive dysfunction, which correspond to structural changes to the medial temporal lobes (MTL) and prefrontal cortex (PFC), respectively. Given the overlap in cognitive deficits between healthy aging and the earliest stages of AD, early detection of AD remains a challenge. The goal of the present study was to study MTL- and PFC-dependent cognitive functioning in middle-aged individuals at genetic risk for AD or cognitive impairment who do not currently manifest any clinical symptoms. Participants (N = 150; aged 40-60 years) underwent genotyping of 47 single nucleotide polymorphisms (SNPs) in six genes previously associated with memory or executive functioning: APOE, SORL1, BDNF, TOMM40, KIBRA, and COMT. They completed two MTL-dependent tasks, the virtual Morris Water Task (vMWT) and transverse patterning discriminations task (TPDT), and the PFC-dependent reversal learning task. Although age was associated with poorer performance on the vMWT and TPDT within this middle-aged sample, there were no genotype-associated differences in cognitive performance. Although the vMWT and TPDT may be sensitive to age-related changes in cognition, carriers of APOE, SORL1, BDNF, TOMM40, KIBRA, and COMT risk alleles do not exhibit alteration in MTL- and PFC-dependent functioning in middle age compared to non-carriers.
Collapse
Affiliation(s)
- Laura E Korthauer
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Elizabeth Awe
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.,Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marijam Frahmand
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Ira Driscoll
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| |
Collapse
|
33
|
Harrington MO, Klaus K, Vaht M, Harro J, Pennington K, Durrant SJ. Overnight retention of emotional memories is influenced by BDNF Val66Met but not 5-HTTLPR. Behav Brain Res 2019; 359:17-27. [DOI: 10.1016/j.bbr.2018.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/24/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023]
|
34
|
Abstract
For more than 50 years, psychologists, gerontologists, and, more recently, neuroscientists have considered the possibility of successful aging. How to define successful aging remains debated, but well-preserved age-sensitive cognitive functions, like episodic memory, is an often-suggested criterion. Evidence for successful memory aging comes from cross-sectional and longitudinal studies showing that some older individuals display high and stable levels of performance. Successful memory aging may be accomplished via multiple paths. One path is through brain maintenance, or relative lack of age-related brain pathology. Through another path, successful memory aging can be accomplished despite brain pathology by means of efficient compensatory and strategic processes. Genetic, epigenetic, and lifestyle factors influence memory aging via both paths. Some of these factors can be promoted throughout the life course, which, at the individual as well as the societal level, can positively impact successful memory aging.
Collapse
Affiliation(s)
- Lars Nyberg
- Department of Radiation Sciences, Umeå University, S-90187 Umeå, Sweden
- Department of Integrative Medical Biology, Umeå University, S-90187 Umeå, Sweden
- Umeå center for Functional Brain Imaging, Umeå University, S-90187 Umeå, Sweden
| | - Sara Pudas
- Department of Integrative Medical Biology, Umeå University, S-90187 Umeå, Sweden
- Umeå center for Functional Brain Imaging, Umeå University, S-90187 Umeå, Sweden
| |
Collapse
|
35
|
Foster CM, Kennedy KM, Horn MM, Hoagey DA, Rodrigue KM. Both hyper- and hypo-activation to cognitive challenge are associated with increased beta-amyloid deposition in healthy aging: A nonlinear effect. Neuroimage 2018; 166:285-292. [PMID: 29108941 DOI: 10.1016/j.neuroimage.2017.10.068] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 11/29/2022] Open
Abstract
Beta-amyloid (Aβ) positive individuals hyper-activate brain regions compared to those not at-risk; however, hyperactivation is then thought to diminish as Alzheimer's disease symptomatology begins, evidencing eventual hypoactivation. It remains unclear when in the disease staging this transition occurs. We hypothesized that differential levels of amyloid burden would be associated with both increased and decreased activation (i.e., a quadratic trajectory) in cognitively-normal adults. Participants (N = 62; aged 51-94) underwent an fMRI spatial distance-judgment task and Amyvid-PET scanning. Voxelwise regression modeled age, linear-Aβ, and quadratic-Aβ as predictors of BOLD activation to difficult spatial distance-judgments. A significant quadratic-Aβ effect on BOLD response explained differential activation in bilateral angular/temporal and medial prefrontal cortices, such that individuals with slightly elevated Aβ burden exhibited hyperactivation whereas even higher Aβ burden was then associated with hypoactivation. Importantly, in high-Aβ individuals, Aβ load moderated the effect of BOLD activation on behavioral task performance, where in lower-elevation, greater deactivation was associated with better accuracy, but in higher-elevation, greater deactivation was associated with poorer accuracy during the task. This study reveals a dose-response, quadratic relationship between increasing Aβ burden and alterations in BOLD activation to cognitive challenge in cognitively-normal individuals that suggests 1) the shift from hyper-to hypo-activation may begin early in disease staging, 2) depends, in part, on degree of Aβ burden, and 3) tracks cognitive performance.
Collapse
Affiliation(s)
- Chris M Foster
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Kristen M Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Marci M Horn
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - David A Hoagey
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Karen M Rodrigue
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA.
| |
Collapse
|
36
|
Tan CJ, Lim SWT, Toh YL, Ng T, Yeo A, Shwe M, Foo KM, Chu P, Jain A, Koo SL, Dent RA, Ng RCH, Yap YS, Lim EH, Loh KWJ, Chay WY, Lee GE, Tan TJY, Beh SY, Wong M, Chan JJ, Khor CC, Ho HK, Chan A. Replication and Meta-analysis of the Association between BDNF Val66Met Polymorphism and Cognitive Impairment in Patients Receiving Chemotherapy. Mol Neurobiol 2018; 56:4741-4750. [PMID: 30382534 PMCID: PMC6647505 DOI: 10.1007/s12035-018-1410-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 10/24/2018] [Indexed: 01/23/2023]
Abstract
Cancer-related cognitive impairment (CRCI) adversely affects cancer patients. We had previously demonstrated that the BDNF Val66Met genetic polymorphism is associated with lower odds of subjective CRCI in the multitasking and verbal ability domains among breast cancer patients receiving chemotherapy. To further assess our previous findings, we evaluated the association of BDNF Val66Met polymorphism with subjective and objective CRCI in a temporally separate cohort of patients and pooled findings from both the original (n = 145) and current (n = 193) cohorts in a meta-analysis. Subjective CRCI was assessed using FACT-Cog. Objective CRCI was evaluated using computerized neuropsychological tests. Genotyping was carried out using Sanger sequencing. The association of BDNF Val66Met genotypes and CRCI was examined with logistic regression. A fixed-effect meta-analysis was conducted using the inverse variance method. In the meta-analysis (n = 338), significantly lower odds of CRCI were associated with Met allele carriers based on the global FACT-Cog score (OR = 0.52, 95% CI 0.29–0.94). Furthermore, Met allele carriers were at lower odds of developing impairment in the domains of memory (OR = 0.34, 95% CI: 0.17–0.70), multitasking (OR = 0.33, 95% CI: 0.18–0.59), and verbal ability (OR = 0.46, 95% CI: 0.24–0.88). Consistent with the previous study, lower odds of subjective CRCI among patients with the BDNF Met allele was observed after adjusting for potential confounders in the multitasking (OR = 0.30, 95% CI: 0.14–0.67) domain. In conclusion, carriers of the BDNF Met allele were protected against global subjective CRCI, particularly in the domains of memory, multitasking, and verbal ability. Our findings further contribute to the understanding of CRCI pathophysiology.
Collapse
Affiliation(s)
- Chia Jie Tan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Sheree Wan Ting Lim
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Yi Long Toh
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Terence Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Angie Yeo
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Maung Shwe
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Koon Mian Foo
- Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore, Singapore
| | - Pat Chu
- Singapore Cord Blood Bank, Singapore, Singapore
| | - Amit Jain
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Si-Lin Koo
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Rebecca A Dent
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | | | - Yoon Sim Yap
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Elaine H Lim
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Kiley Wei-Jen Loh
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Wen Yee Chay
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Guek Eng Lee
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Tira Jing Ying Tan
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Sok Yuen Beh
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Mabel Wong
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Jack Junjie Chan
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Chiea Chuen Khor
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore.,Glaucoma Research Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Alexandre Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore. .,Department of Pharmacy, National Cancer Centre, Singapore, Singapore. .,Duke-NUS Graduate Medical School, Singapore, Singapore.
| |
Collapse
|
37
|
Daugherty AM, Hoagey DA, Kennedy KM, Rodrigue KM. Genetic predisposition for inflammation exacerbates effects of striatal iron content on cognitive switching ability in healthy aging. Neuroimage 2018; 185:471-478. [PMID: 30395929 DOI: 10.1016/j.neuroimage.2018.10.064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/25/2018] [Accepted: 10/24/2018] [Indexed: 01/04/2023] Open
Abstract
Non-heme iron homeostasis interacts with inflammation bidirectionally, and both contribute to age-related decline in brain structure and function via oxidative stress. Thus, individuals with genetic predisposition for inflammation may be at greater risk for brain iron accumulation during aging and more vulnerable to cognitive decline. We examine this hypothesis in a lifespan sample of healthy adults (N = 183, age 20-94 years) who underwent R2*-weighted magnetic resonance imaging to estimate regional iron content and genotyping of interleukin-1beta (IL-1β), a pro-inflammatory cytokine for which the T allelle of the single nucleotide polymorphism increases risk for chronic neuroinflammation. Older age was associated with greater striatal iron content that in turn accounted for poorer cognitive switching performance. Heterozygote IL-1β T-carriers demonstrated poorer switching performance in relation to striatal iron content as compared to IL-1β C/C counterparts, despite the two groups being of similar age. With increasing genetic inflammation risk, homozygote IL-1β T/T carriers had lesser age-related variance in striatal iron content as compared to the other groups but showed a similar association of greater striatal iron content predicting poorer cognitive switching. Non-heme iron and inflammation, although necessary for normal neuronal function, both promote oxidative stress that when accumulated in excess, drives a complex mechanism of neural and cognitive decline in aging.
Collapse
Affiliation(s)
- Ana M Daugherty
- Department of Psychology, Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI, USA
| | - David A Hoagey
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Kristen M Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Karen M Rodrigue
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA.
| |
Collapse
|
38
|
Barha CK, Liu-Ambrose T, Best JR, Yaffe K, Rosano C. Sex-dependent effect of the BDNF Val66Met polymorphism on executive functioning and processing speed in older adults: evidence from the health ABC study. Neurobiol Aging 2018; 74:161-170. [PMID: 30448615 DOI: 10.1016/j.neurobiolaging.2018.10.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 02/07/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) Val66Met polymorphism may be an important source of heterogeneity seen in cognitive aging, although the specific relationship between this polymorphism and cognition remains controversial and may depend on the sex of participants. We assessed 2668 older black and white adults and fit linear mixed models to digit symbol substitution test (DSST) performance assessed in years 0 (baseline), 4, 7, and 9 to examine the interaction between sex and BDNF genotype on the intercept (i.e., estimated baseline DSST) and change in DSST over 9 years, adjusted for covariates. Sex interacted with BDNF genotype to predict DSST intercept (F[1,1599] = 7.4, p < 0.01) and 9-year change (F[1,1183] = 4.1, p = 0.04) in white participants only. Initially, white male Val/Val carriers had lower DSST scores (37.6, SE = 0.8) in comparison with male Met carriers (difference, -1.7; 95% CI, -3.2 to -0.3) and female Val/Val carriers (difference, -5.6; 95% CI, -6.8 to -4.3). White female Met carriers showed a slower rate of change (annual rate of change = -0.6, SE = 0.1) in comparison with female Val/Val carriers (difference, -0.2; 95% CI, -0.4 to -0.02) and male Met carriers (difference, -0.3; 95% CI, -0.5 to -0.02). Our findings suggest that BDNF Val66Met and sex should be considered in future endeavors aimed at treating or preventing neurodegenerative disorders.
Collapse
Affiliation(s)
- Cindy K Barha
- Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute, Vancouver, Canada; Department of Physical Therapy, University of British Columbia, Vancouver, Canada.
| | - Teresa Liu-Ambrose
- Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute, Vancouver, Canada; Department of Physical Therapy, University of British Columbia, Vancouver, Canada; Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - John R Best
- Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute, Vancouver, Canada; Department of Physical Therapy, University of British Columbia, Vancouver, Canada
| | - Kristine Yaffe
- Department of Epidemiology and Biostatistics, University of California, San Francisco, USA; Departments of Psychiatry and Neurology, University of California, San Francisco, USA
| | - Caterina Rosano
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, USA
| | | |
Collapse
|
39
|
Rodríguez-Rojo IC, Cuesta P, López ME, de Frutos-Lucas J, Bruña R, Pereda E, Barabash A, Montejo P, Montenegro-Peña M, Marcos A, López-Higes R, Fernández A, Maestú F. BDNF Val66Met Polymorphism and Gamma Band Disruption in Resting State Brain Functional Connectivity: A Magnetoencephalography Study in Cognitively Intact Older Females. Front Neurosci 2018; 12:684. [PMID: 30333719 PMCID: PMC6176075 DOI: 10.3389/fnins.2018.00684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/11/2018] [Indexed: 11/13/2022] Open
Abstract
The pathophysiological processes undermining brain functioning decades before the onset of the clinical symptoms associated with dementia are still not well understood. Several heritability studies have reported that the Brain Derived Neurotrophic Factor (BDNF) Val66Met genetic polymorphism could contribute to the acceleration of cognitive decline in aging. This mutation may affect brain functional connectivity (FC), especially in those who are carriers of the BDNF Met allele. The aim of this work was to explore the influence of the BDNF Val66Met polymorphism in whole brain eyes-closed, resting-state magnetoencephalography (MEG) FC in a sample of 36 cognitively intact (CI) older females. All of them were ε3ε3 homozygotes for the apolipoprotein E (APOE) gene and were divided into two subgroups according to the presence of the Met allele: Val/Met group (n = 16) and Val/Val group (n = 20). They did not differ in age, years of education, Mini-Mental State Examination scores, or normalized hippocampal volumes. Our results showed reduced antero-posterior gamma band FC within the Val/Met genetic risk group, which may be caused by a GABAergic network impairment. Despite the lack of cognitive decline, these results might suggest a selective brain network vulnerability due to the carriage of the BDNF Met allele, which is linked to a potential progression to dementia. This neurophysiological signature, as tracked with MEG FC, indicates that age-related brain functioning changes could be mediated by the influence of particular genetic risk factors.
Collapse
Affiliation(s)
- Inmaculada C Rodríguez-Rojo
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.,Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Pablo Cuesta
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.,Electrical Engineering and Bioengineering Lab, Department of Industrial Engineering and IUNE, Universidad de La Laguna, Tenerife, Spain
| | - María Eugenia López
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.,Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Jaisalmer de Frutos-Lucas
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.,Biological and Health Psychology Department, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ricardo Bruña
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.,Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Ernesto Pereda
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.,Electrical Engineering and Bioengineering Lab, Department of Industrial Engineering and IUNE, Universidad de La Laguna, Tenerife, Spain
| | - Ana Barabash
- Laboratory of Psychoneuroendocrinology and Genetics, Hospital Clínico San Carlos, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Pedro Montejo
- Center for the Prevention of Cognitive Impairment, Public Health Institute, Madrid-Salud, Madrid, Spain
| | - Mercedes Montenegro-Peña
- Center for the Prevention of Cognitive Impairment, Public Health Institute, Madrid-Salud, Madrid, Spain
| | - Alberto Marcos
- Neurology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Ramón López-Higes
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Alberto Fernández
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.,Department of Legal Medicine, Psychiatry, and Pathology, Universidad Complutense de Madrid, Madrid, Spain
| | - Fernando Maestú
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.,Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
40
|
Jacobs EG, Goldstein JM. The Middle-Aged Brain: Biological sex and sex hormones shape memory circuitry. Curr Opin Behav Sci 2018; 23:84-91. [PMID: 30271832 PMCID: PMC6157917 DOI: 10.1016/j.cobeha.2018.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Emily G. Jacobs
- Department of Psychological and Brain Sciences, University of California, Santa Barbara
- Neuroscience Research Institute, University of California, Santa Barbara
| | - Jill M. Goldstein
- Departments of Psychiatry and Medicine, Harvard Medical School
- Departments of Psychiatry and Obstetrics and Gynecology, Massachusetts General Hospital
- Athinoula A. Martinos Brain Imaging Center, Massachusetts General Hospital
| |
Collapse
|
41
|
Balkaya M, Cho S. Genetics of stroke recovery: BDNF val66met polymorphism in stroke recovery and its interaction with aging. Neurobiol Dis 2018; 126:36-46. [PMID: 30118755 DOI: 10.1016/j.nbd.2018.08.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/24/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke leads to long term sensory, motor and cognitive impairments. Most patients experience some degree of spontaneous recovery which is mostly incomplete and varying greatly among individuals. The variation in recovery outcomes has been attributed to numerous factors including lesion size, corticospinal tract integrity, age, gender and race. It is well accepted that genetics play a crucial role in stroke incidence and accumulating evidence suggests that it is also a significant determinant in recovery. Among the number of genes and variations implicated in stroke recovery the val66met single nucleotide polymorphism (SNP) in the BDNF gene influences post-stroke plasticity in the most significant ways. Val66met is the most well characterized BDNF SNP and is common (40-50 % in Asian and 25-32% in Caucasian populations) in humans. It reduces activity-dependent BDNF release, dampens cortical plasticity and is implicated in numerous diseases. Earlier studies on the effects of val66met on stroke outcome and recovery presented primarily a maladaptive role. Novel findings however indicate a much more intricate interaction between val66met and stroke recovery which appears to be influenced by lesion location, post-stroke stage and age. This review will focus on the role of BDNF and val66met SNP in relation to stroke recovery and try to identify potential pathophysiologic mechanisms involved. The effects of age on val66met associated alterations in plasticity and potential consequences in terms of stroke are also discussed.
Collapse
Affiliation(s)
- Mustafa Balkaya
- Burke-Cornell Medical Research Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine at Burke Medical Research Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA
| | - Sunghee Cho
- Burke-Cornell Medical Research Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine at Burke Medical Research Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA.
| |
Collapse
|
42
|
Toh YL, Ng T, Tan M, Tan A, Chan A. Impact of brain-derived neurotrophic factor genetic polymorphism on cognition: A systematic review. Brain Behav 2018; 8:e01009. [PMID: 29858545 PMCID: PMC6043712 DOI: 10.1002/brb3.1009] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 04/08/2018] [Accepted: 04/15/2018] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Brain-derived neurotrophic factor (BDNF) has an important role in the neurogenesis and neuroplasticity of the brain. This systematic review was designed to examine the association between BDNF Val66Met (rs6265) polymorphism and four cognitive domains-attention and concentration, executive function, verbal fluency, and memory, respectively. METHODOLOGY Primary literature search was performed using search engines such as PubMed and Scopus. Observational studies that evaluated the neurocognitive performances in relation to BDNF polymorphism within human subjects were included in this review, while animal studies, overlapping studies, and meta-analysis were excluded. RESULTS Forty of 82 reviewed studies (48.8%) reported an association between Val66Met polymorphism and neurocognitive domains. The proportion of the studies showing positive findings in cognitive performances between Val/Val homozygotes and Met carriers was comparable, at 30.5% and 18.3%, respectively. The highest percentage of positive association between Val66Met polymorphism and neurocognition was reported under the memory domain, with 26 of 63 studies (41.3%), followed by 18 of 47 studies (38.3%) under the executive function domain and four of 23 studies (17.4%) under the attention and concentration domain. There were no studies showing an association between Val66Met polymorphism and verbal fluency. In particular, Val/Val homozygotes performed better in tasks related to the memory domain, while Met carriers performed better in terms of executive function, in both healthy individuals and clinical populations. CONCLUSION While numerous studies report an association between Val66Met polymorphism and neurocognitive changes in executive function and memory domains, the effect of Met allele has not been clearly established.
Collapse
Affiliation(s)
- Yi Long Toh
- Department of PharmacyFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Terence Ng
- Department of PharmacyFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Megan Tan
- Department of PharmacyFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Azrina Tan
- Department of PharmacyFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Alexandre Chan
- Department of PharmacyFaculty of ScienceNational University of SingaporeSingaporeSingapore
- Department of PharmacyNational Cancer Centre SingaporeSingaporeSingapore
| |
Collapse
|
43
|
Horn MM, Kennedy KM, Rodrigue KM. Association between subjective memory assessment and associative memory performance: Role of ad risk factors. Psychol Aging 2018; 33:109-118. [PMID: 29494182 PMCID: PMC5836750 DOI: 10.1037/pag0000217] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Decline in associative memory abilities is a common cognitive complaint among older adults and is detectable in both normal aging and in prodromal Alzheimer's disease (AD). Subjective memory (SM) complaints may serve as an earlier marker of these mnemonic changes; however, previous research examining the predictive utility of SM to observed memory performance yielded inconsistent results. This inconsistency is likely due to other sources of variance that occur with memory decline such as mood/depression issues, presence of apolipoprotein E (APOE ε4) genotype, or beta-amyloid deposition. Here we examine the relationship between SM and associative memory ability in the context of factors that increase susceptibility to AD in 195 healthy adults (79 men) aged 20-94 years. Participants completed an SM questionnaire, a mood/depression scale, two associative memory tests (a word-pair and a name-face test), and were genotyped for APOE ε4. PET-amyloid imaging data were collected for a subset of those over 50 years of age (N = 74). We found that SM predicted performance on both associative memory tests even after covarying for age, sex, mood, and APOE ε4 status. Interestingly, for the name-face associative task, increased SM concerns predicted memory performance selectively in participants over the age of 60, with the APOEε4 risk group showing the strongest effect. Finally, men with higher beta-amyloid deposition reported more memory complaints. Our findings suggest that SM reliably tracks memory performance, even in cognitively healthy adults, and may reflect an increased risk for AD. (PsycINFO Database Record
Collapse
Affiliation(s)
- Marci M. Horn
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235
| | - Kristen M. Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235
| | - Karen M. Rodrigue
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235
| |
Collapse
|
44
|
Porter T, Burnham SC, Doré V, Savage G, Bourgeat P, Begemann K, Milicic L, Ames D, Bush AI, Maruff P, Masters CL, Rowe CC, Rainey-Smith S, Martins RN, Groth D, Verdile G, Villemagne VL, Laws SM. KIBRA is associated with accelerated cognitive decline and hippocampal atrophy in APOE ε4-positive cognitively normal adults with high Aβ-amyloid burden. Sci Rep 2018; 8:2034. [PMID: 29391469 PMCID: PMC5794989 DOI: 10.1038/s41598-018-20513-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/19/2018] [Indexed: 12/26/2022] Open
Abstract
A single nucleotide polymorphism, rs17070145, in the KIdney and BRAin expressed protein (KIBRA) gene has been associated with cognition and hippocampal volume in cognitively normal (CN) individuals. However, the impact of rs17070145 on longitudinal cognitive decline and hippocampal atrophy in CN adults at greatest risk of developing Alzheimer's disease is unknown. We investigated the impact rs17070145 has on the rate of cognitive decline and hippocampal atrophy over six years in 602 CN adults, with known brain Aβ-amyloid levels and whether there is an interactive effect with APOE genotype. We reveal that whilst limited independent effects of KIBRA genotype were observed, there was an interaction with APOE in CN adults who presented with high Aβ-amyloid levels across study duration. In comparison to APOE ε4-ve individuals carrying the rs17070145-T allele, significantly faster rates of cognitive decline (global, p = 0.006; verbal episodic memory, p = 0.004), and hippocampal atrophy (p = 0.04) were observed in individuals who were APOE ε4 + ve and did not carry the rs17070145-T allele. The observation of APOE effects in only non-carriers of the rs17070145-T allele, in the presence of high Aβ-amyloid suggest that carriers of the rs17070145-T allele are conferred a level of resilience to the detrimental effects of high Aβ-amyloid and APOE ε4.
Collapse
Affiliation(s)
- Tenielle Porter
- Collaborative Genomics Group, Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, 6027, Western Australia, Australia.,Co-operative Research Centre for Mental Health, Carlton South, 3053 Victoria, Australia
| | - Samantha C Burnham
- CSIRO Health and Biosecurity, Parkville, 3052, Victoria, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, 6027, Western Australia, Australia
| | - Vincent Doré
- eHealth, CSIRO Health and Biosecurity, Herston, 4029, QLD, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, 3084, Victoria, Australia
| | - Greg Savage
- ARC Centre of Excellence in Cognition and its Disorders, Department of Psychology, Macquarie University, North Ryde, 2113, NSW, Australia
| | - Pierrick Bourgeat
- eHealth, CSIRO Health and Biosecurity, Herston, 4029, QLD, Australia
| | - Kimberly Begemann
- Collaborative Genomics Group, Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, 6027, Western Australia, Australia
| | - Lidija Milicic
- Collaborative Genomics Group, Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, 6027, Western Australia, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, St. Vincent's Health, The University of Melbourne, Kew, 3101, Victoria, Australia.,National Ageing Research Institute, Parkville, 3052, Victoria, Australia
| | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, 3052, Victoria, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, 3052, Victoria, Australia.,CogState Ltd., Melbourne, 3000, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, 3052, Victoria, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, 3084, Victoria, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, 3084, Victoria, Australia
| | - Stephanie Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, 6027, Western Australia, Australia
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, 6027, Western Australia, Australia.,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, 6009, Western Australia, Australia
| | - David Groth
- School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
| | - Giuseppe Verdile
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, 6027, Western Australia, Australia.,School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
| | - Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, 3084, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, 3052, Victoria, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, 3084, Victoria, Australia
| | - Simon M Laws
- Collaborative Genomics Group, Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, 6027, Western Australia, Australia. .,Co-operative Research Centre for Mental Health, Carlton South, 3053 Victoria, Australia. .,School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia.
| |
Collapse
|
45
|
Azeredo LA, De Nardi T, Grassi-Oliveira R. BDNF Val66Met polymorphism and memory performance in older adults: the Met carrier effect is more complex than previously thought: Authors’ reply. REVISTA BRASILEIRA DE PSIQUIATRIA 2017; 39:276-277. [PMID: 28813118 PMCID: PMC7111378 DOI: 10.1590/1516-4446-2017-3904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 11/22/2022]
Affiliation(s)
| | | | - Rodrigo Grassi-Oliveira
- Pontifícia Universidade Católica do Rio Grande do Sul, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Brazil
| |
Collapse
|
46
|
BDNF Val66Met Polymorphism Interacts with Sleep Consolidation to Predict Ability to Create New Declarative Memories. J Neurosci 2017; 36:8390-8. [PMID: 27511011 DOI: 10.1523/jneurosci.4432-15.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 06/23/2016] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED It is hypothesized that a fundamental function of sleep is to restore an individual's day-to-day ability to learn and to constantly adapt to a changing environment through brain plasticity. Brain-derived neurotrophic factor (BDNF) is among the key regulators that shape brain plasticity. However, advancing age and carrying the BDNF Met allele were both identified as factors that potentially reduce BDNF secretion, brain plasticity, and memory. Here, we investigated the moderating role of BDNF polymorphism on sleep and next-morning learning ability in 107 nondemented individuals who were between 55 and 84 years of age. All subjects were tested with 1 night of in-laboratory polysomnography followed by a cognitive evaluation the next morning. We found that in subjects carrying the BDNF Val66Val polymorphism, consolidated sleep was associated with significantly better performance on hippocampus-dependent episodic memory tasks the next morning (β-values from 0.290 to 0.434, p ≤ 0.01). In subjects carrying at least one copy of the BDNF Met allele, a more consolidated sleep was not associated with better memory performance in most memory tests (β-values from -0.309 to -0.392, p values from 0.06 to 0.15). Strikingly, increased sleep consolidation was associated with poorer performance in learning a short story presented verbally in Met allele carriers (β = -0.585, p = 0.005). This study provides new evidence regarding the interacting roles of consolidated sleep and BDNF polymorphism in the ability to learn and stresses the importance of considering BDNF polymorphism when studying how sleep affects cognition. SIGNIFICANCE STATEMENT Individuals with the BDNF Val/Val (valine allele) polymorphism showed better memory performance after a night of consolidated sleep. However, we observed that middle-aged and older individuals who are carriers of the BDNF Met allele displayed no positive association between sleep quality and their ability to learn the next morning. This interaction between sleep and BDNF polymorphism was more salient for hippocampus-dependent tasks than for other cognitive tasks. Our results support the hypothesis that reduced activity-dependent secretion of BDNF impairs the benefits of sleep on synaptic plasticity and next-day memory. Our work advances the field by revealing new evidence of a clear genetic heterogeneity in how sleep consolidation contributes to the ability to learn.
Collapse
|
47
|
Differential Aging Trajectories of Modulation of Activation to Cognitive Challenge in APOE ε4 Groups: Reduced Modulation Predicts Poorer Cognitive Performance. J Neurosci 2017; 37:6894-6901. [PMID: 28652414 DOI: 10.1523/jneurosci.3900-16.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 06/01/2017] [Accepted: 06/08/2017] [Indexed: 01/08/2023] Open
Abstract
The present study was designed to investigate the effect of a genetic risk factor for Alzheimer's disease (AD), ApolipoproteinE ε4 (APOEε4), on the ability of the brain to modulate activation in response to cognitive challenge in a lifespan sample of healthy human adults. A community-based sample of 181 cognitively intact, healthy adults were recruited from the Dallas-Fort Worth metroplex. Thirty-one APOEε4+ individuals (48% women), derived from the parent sample, were matched based on sex, age, and years of education to 31 individuals who were APOEε4-negative (APOEε4-). Ages ranged from 20 to 86 years of age. Blood oxygen level-dependent functional magnetic resonance imaging was collected during the performance of a visuospatial distance judgment task with three parametric levels of difficulty. Multiple regression was used in a whole-brain analysis with age, APOE group, and their interaction predicting functional brain modulation in response to difficulty. Results revealed an interaction between age and APOE in a large cluster localized primarily to the bilateral precuneus. APOEε4- individuals exhibited age-invariant modulation in response to task difficulty, whereas APOEε4+ individuals showed age-related reduction of modulation in response to increasing task difficulty compared with ε4- individuals. Decreased modulation in response to cognitive challenge was associated with reduced task accuracy as well as poorer name-face associative memory performance. Findings suggest that APOEε4 is associated with a reduction in the ability of the brain to dynamically modulate in response to cognitive challenge. Coupled with a significant genetic risk factor for AD, changes in modulation may provide additional information toward identifying individuals potentially at risk for cognitive decline associated with preclinical AD.SIGNIFICANCE STATEMENT Understanding how risk factors for Alzheimer's disease (AD) affect brain function and cognition in healthy adult samples may help to identify the biomarkers needed to detect nonsymptomatic, preclinical phases of the disease. Findings from the current study show that ApolipoproteinE ε4-positive (APOEε4+) individuals exhibit an altered lifespan trajectory in the ability of the brain to dynamically modulate function to cognitive challenge compared with APOEε4- individuals. This effect manifests in otherwise healthy individuals who are at increased risk for AD in the precuneus, a salient region for early AD changes. Notably, these functional alterations are detrimental to performance, and thus, the combination of a genetic risk factor and altered modulation may provide important information for identifying individuals who are at increased risk for AD.
Collapse
|
48
|
Avgan N, Sutherland HG, Lea RA, Spriggens LK, Haupt LM, Shum DHK, Griffiths LR. A CREB1 Gene Polymorphism (rs2253206) Is Associated with Prospective Memory in a Healthy Cohort. Front Behav Neurosci 2017; 11:86. [PMID: 28559802 PMCID: PMC5432533 DOI: 10.3389/fnbeh.2017.00086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/25/2017] [Indexed: 11/18/2022] Open
Abstract
Prospective memory (PM) is generally defined as remembering to perform intended actions in the future and is important for functioning in daily life. Cyclic adenosine monophosphate (cAMP) responsive element binding protein 1 (CREB1) plays an important role in cognitive functions. In this study, we hypothesized that genetic variation in the CREB1 gene is associated with PM. We genotyped a CREB1 promoter polymorphism rs2253206 and tested it for association with PM in 619 healthy subjects. PM performance was measured using the Prospective and Retrospective Memory Questionnaire (PRMQ), the Comprehensive Assessment of Prospective Memory (CAPM), and the Memory for Intentions Screening Test (MIST). Generalized linear model analysis was conducted for each PM test independently using different inheritance models to identify any associations (p < 0.05). After multiple testing adjustments, a significant association was found between the rs2253206 genotype and PM performance for CAPM instrumental activities of daily living measure (p = 0.016). These results suggest that the rs2253206 polymorphism in the CREB1 gene locus is associated with PM in healthy individuals and contributes to knowledge on the genetics of this particular type of memory.
Collapse
Affiliation(s)
- Nesli Avgan
- Genomics Research Centre, Chronic Disease and Ageing, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, BrisbaneQLD, Australia
| | - Heidi G Sutherland
- Genomics Research Centre, Chronic Disease and Ageing, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, BrisbaneQLD, Australia
| | - Rodney A Lea
- Genomics Research Centre, Chronic Disease and Ageing, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, BrisbaneQLD, Australia
| | - Lauren K Spriggens
- Menzies Health Institute Queensland and School of Applied Psychology, Griffith University, Gold CoastQLD, Australia
| | - Larisa M Haupt
- Genomics Research Centre, Chronic Disease and Ageing, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, BrisbaneQLD, Australia
| | - David H K Shum
- Menzies Health Institute Queensland and School of Applied Psychology, Griffith University, Gold CoastQLD, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Chronic Disease and Ageing, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, BrisbaneQLD, Australia
| |
Collapse
|
49
|
Ward DD, Andel R, Saunders NL, Thow ME, Klekociuk SZ, Bindoff AD, Vickers JC. The BDNF Val66Met polymorphism moderates the effect of cognitive reserve on 36-month cognitive change in healthy older adults. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:323-331. [PMID: 29067339 PMCID: PMC5651414 DOI: 10.1016/j.trci.2017.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction Cognitive reserve (CR) and BDNF Val66Met are independently associated with the rate of cognitive decline in preclinical Alzheimer's disease. This study was designed to investigate the interactive effects of these variables on 36-month cognitive change in cognitively intact older adults. Methods Data for this investigation were obtained from 445 community-residing participants of the Tasmanian Healthy Brain Project, who underwent genetic screening and annual assessment of neuropsychological, health, and psychosocial function. Results Our main result was that BDNF Val66Met moderated the relationship between baseline CR and change in executive function performance, in that CR-related differences in function decreased across the follow-up period in BDNF Val homozygotes, but became more pronounced in BDNF Met carriers. Similar effects were not observed within the other memory- and language-related cognitive domains. Discussion Inheritance of BDNF Met may be associated with a detrimental influence on the relationship between CR and cognitive change in cognitively intact older adults, but this effect may be restricted to the executive function domain.
Collapse
Affiliation(s)
- David D Ward
- Wicking Dementia Research & Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Ross Andel
- School of Aging Studies, University of South Florida, Tampa, FL, USA
| | - Nichole L Saunders
- Wicking Dementia Research & Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Megan E Thow
- Wicking Dementia Research & Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Shannon Z Klekociuk
- Wicking Dementia Research & Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Aidan D Bindoff
- Wicking Dementia Research & Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research & Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
50
|
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: 47] [Impact Index Per Article: 5.9] [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
|