1
|
Treble-Barna A, Petersen BA, Stec Z, Conley YP, Fink EL, Kochanek PM. Brain-Derived Neurotrophic Factor in Pediatric Acquired Brain Injury and Recovery. Biomolecules 2024; 14:191. [PMID: 38397427 PMCID: PMC10886547 DOI: 10.3390/biom14020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
We review emerging preclinical and clinical evidence regarding brain-derived neurotrophic factor (BDNF) protein, genotype, and DNA methylation (DNAm) as biomarkers of outcomes in three important etiologies of pediatric acquired brain injury (ABI), traumatic brain injury, global cerebral ischemia, and stroke. We also summarize evidence suggesting that BDNF is (1) involved in the biological embedding of the psychosocial environment, (2) responsive to rehabilitative therapies, and (3) potentially modifiable. BDNF's unique potential as a biomarker of neuroplasticity and neural repair that is reflective of and responsive to both pre- and post-injury environmental influences separates it from traditional protein biomarkers of structural brain injury with exciting potential to advance pediatric ABI management by increasing the accuracy of prognostic tools and informing clinical decision making through the monitoring of therapeutic effects.
Collapse
Affiliation(s)
- Amery Treble-Barna
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
| | - Bailey A. Petersen
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
| | - Zachary Stec
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
| | - Yvette P. Conley
- Department of Health Promotion & Development, University of Pittsburgh School of Nursing, Pittsburgh, PA 15213, USA;
| | - Ericka L. Fink
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
2
|
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
|
3
|
Galindo C, Nguyen VT, Hill B, Sims N, Heck A, Negron M, Lusk C. Brain-derived neurotrophic factor rs6265 (Val66Met) single nucleotide polymorphism as a master modifier of human pathophysiology. Neural Regen Res 2023. [PMID: 35799516 PMCID: PMC9241394 DOI: 10.4103/1673-5374.343894] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Brain-derived neurotrophic factor is the most prevalent member of the nerve growth factor family. Since its discovery in 1978, this enigmatic molecule has spawned more than 27,000 publications, most of which are focused on neurological disorders. Brain-derived neurotrophic factor is indispensable during embryogenesis and postnatally for the normal development and function of both the central and peripheral nervous systems. It is becoming increasingly clear, however, that brain-derived neurotrophic factor likewise plays crucial roles in a variety of other biological functions independently of sympathetic or parasympathetic involvement. Brain-derived neurotrophic factor is also increasingly recognized as a sophisticated environmental sensor and master coordinator of whole organismal physiology. To that point, we recently found that a common nonsynonymous (Val66→Met) single nucleotide polymorphism in the brain-derived neurotrophic factor gene (rs6265) not only substantially alters basal cardiac transcriptomics in mice but subtly influences heart gene expression and function differentially in males and females. In addition to a short description of recent results from associative neuropsychiatric studies, this review provides an eclectic assortment of research reports that support a modulatory role for rs6265 including and beyond the central nervous system.
Collapse
|
4
|
Azman KF, Zakaria R. Recent Advances on the Role of Brain-Derived Neurotrophic Factor (BDNF) in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:6827. [PMID: 35743271 PMCID: PMC9224343 DOI: 10.3390/ijms23126827] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
Neurotrophins, such as brain-derived neurotrophic factor (BDNF), are essential for neuronal survival and growth. The signaling cascades initiated by BDNF and its receptor are the key regulators of synaptic plasticity, which plays important role in learning and memory formation. Changes in BDNF levels and signaling pathways have been identified in several neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease, and have been linked with the symptoms and course of these diseases. This review summarizes the current understanding of the role of BDNF in several neurodegenerative diseases, as well as the underlying molecular mechanism. The therapeutic potential of BDNF treatment is also discussed, in the hope of discovering new avenues for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Khairunnuur Fairuz Azman
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
| | | |
Collapse
|
5
|
Khodir SA, Faried MA, Abd-Elhafiz HI, Sweed EM. Sitagliptin Attenuates the Cognitive Deficits in L-Methionine-Induced Vascular Dementia in Rats. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7222590. [PMID: 35265716 PMCID: PMC8898801 DOI: 10.1155/2022/7222590] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/29/2022] [Indexed: 02/05/2023]
Abstract
Vascular dementia (VaD) is the second most prevalent type of dementia characterized by progressive cognitive deficits and is a major risk factor for the development of Alzheimer's disease and other neurodegenerative disorders. This study is aimed at determining the potential neuroprotective effect of sitagliptin (STG) on cognitive deficits in L-methionine-induced VaD in rats and the possible underlying mechanisms. 30 adult male Wistar albino rats were divided equally (n = 10) into three groups: control, VaD, and VaD + STG groups. The cognitive performance of the animals was conducted by open field, elevated plus maze, Y-maze, novel object recognition, and Morris water maze tests. Serum homocysteine, TNF-α, IL-6, IL-10, total cholesterol, and triglycerides levels were assessed together with hippocampal MDA, SOD, and BDNF. Histopathological and immunohistochemical assessments of the thoracic aorta and hippocampus (CA1 region) were also performed. Chronic L-methionine administration impaired memory and learning and induced anxiety. On the other hand, STG protected against cognitive deficits through improving oxidative stress biomarkers, inflammatory mediators, lipid profiles, and hippocampus level of BDNF as well as decreasing caspase-3 and GFAP and increasing Ki-67 immunoreactions in the hippocampus. Also, STG improved the endothelial dysfunction via upregulation of aortic eNOS immunoreaction. STG improved the cognitive deficits of L-methionine-induced VaD by its antioxidant, anti-inflammatory, antiapoptotic, and neurotrophic effects. These findings suggest that STG may be a promising future agent for protection against VaD.
Collapse
Affiliation(s)
- Suzan A. Khodir
- 1Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Manar A. Faried
- 2Human Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Huda I. Abd-Elhafiz
- 3Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Eman M. Sweed
- 3Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| |
Collapse
|
6
|
Chung J, Ushakova A, Doitsidou M, Tzoulis C, Tysnes OB, Dalen I, Pedersen KF, Alves G, Maple-Grødem J. The impact of common genetic variants in cognitive decline in the first seven years of Parkinson's disease: A longitudinal observational study. Neurosci Lett 2021; 764:136243. [PMID: 34509566 DOI: 10.1016/j.neulet.2021.136243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Cognitive impairment is a common feature of Parkinson's disease and is a significant determinant of patients' quality of life and dependence. The pattern and progression of cognitive symptoms vary greatly between individuals, and genetic biomarkers may help to predict the severity and trajectory of cognitive impairment in groups of patients. METHODS The study included 171 patients from a longitudinal population-based incident Parkinson's disease study from South Western Norway. All participants were followed from the time of diagnosis for up to seven years, undertaking repeated batteries of clinical and neuropsychological tests, measuring global cognitive impairment, executive function, attention, verbal learning and memory, and visuospatial skills. We used linear mixed regression analyses to explore associations between the function in specific cognitive domains over time and common genetic variants in APOE, MAPT, COMT and BDNF. RESULTS The COMT158Val/Val allele wasassociatedwith faster decline in executive function (p = 0.028), verbal learning and memory (p = 0.029), and visuospatial skills (p = 0.027). The BDNF, MAPT and APOE genotypes were not significantly associated with longitudinal changes in individual cognitive domains, however carriers of the APOE-ε4 allele were shown to be at increased risk of mild cognitive impairment and dementia within the study period (OR3.03; p = 0.006). CONCLUSIONS This population-based study of newly diagnosed patients provides new evidence that COMTVal158Met effects cognitive outcomes limited to discrete domains and APOE-ε4 status predicts a poor overall cognitive prognosis. Together, these data contribute to our understanding of the biology underlying the heterogeneity observed in the progression of PD.
Collapse
Affiliation(s)
- Janete Chung
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
| | - Anastasia Ushakova
- Department of Research, Section of Biostatistics, Stavanger University Hospital, Stavanger, Norway
| | - Maria Doitsidou
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | | | - Ole-Bjørn Tysnes
- Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Norway
| | - Ingvild Dalen
- Department of Research, Section of Biostatistics, Stavanger University Hospital, Stavanger, Norway
| | - Kenn Freddy Pedersen
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Guido Alves
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Neurology, Stavanger University Hospital, Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Jodi Maple-Grødem
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.
| |
Collapse
|
7
|
Gonzalez-Latapi P, Bayram E, Litvan I, Marras C. Cognitive Impairment in Parkinson's Disease: Epidemiology, Clinical Profile, Protective and Risk Factors. Behav Sci (Basel) 2021; 11:bs11050074. [PMID: 34068064 PMCID: PMC8152515 DOI: 10.3390/bs11050074] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023] Open
Abstract
Cognitive impairment is a common non-motor symptom in Parkinson's Disease (PD) and an important source of patient disability and caregiver burden. The timing, profile and rate of cognitive decline varies widely among individuals with PD and can range from normal cognition to mild cognitive impairment (PD-MCI) and dementia (PDD). Beta-amyloid and tau brain accumulation, oxidative stress and neuroinflammation are reported risk factors for cognitive impairment. Traumatic brain injury and pesticide and tobacco exposure have also been described. Genetic risk factors including genes such as COMT, APOE, MAPT and BDNF may also play a role. Less is known about protective factors, although the Mediterranean diet and exercise may fall in this category. Nonetheless, there is conflicting evidence for most of the factors that have been studied. The use of inconsistent criteria and lack of comprehensive assessment in many studies are important methodological issues. Timing of exposure also plays a crucial role, although identification of the correct time window has been historically difficult in PD. Our understanding of the mechanism behind these factors, as well as the interactions between gene and environment as determinants of disease phenotype and the identification of modifiable risk factors will be paramount, as this will allow for potential interventions even in established PD.
Collapse
Affiliation(s)
- Paulina Gonzalez-Latapi
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Toronto, ON M5T2S8, Canada;
| | - Ece Bayram
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; (E.B.); (I.L.)
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; (E.B.); (I.L.)
| | - Connie Marras
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Toronto, ON M5T2S8, Canada;
- Correspondence:
| |
Collapse
|
8
|
Oberste M, Sharma S, Bloch W, Zimmer P. Acute Exercise-Induced Set Shifting Benefits in Healthy Adults and Its Moderators: A Systematic Review and Meta-Analysis. Front Psychol 2021; 12:528352. [PMID: 33584460 PMCID: PMC7879782 DOI: 10.3389/fpsyg.2021.528352] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Positive effects of acute exercise on cognitive performances in general inspired research that investigated the effects of acute exercise on specific cognitive subdomains. Many existing studies examined beneficial effects of acute exercise on subsequent set shifting performance in healthy adults. Set shifting, a subdomain of executive function, is the ability to switch between different cognitive sets. The results of existing studies are inconsistent. Therefore, a meta-analysis was conducted that pooled available effect sizes. Additionally, moderator analyses were carried out to identify covariates that determine the magnitude of exercise-induced set shifting benefits. Methods: Medline, PsycINFO, and SPORTDiscus were searched for eligible studies. Hedges' g corrected standardized mean difference values were used for analyses. Random-effects weights were applied to pool effects. Potential moderation of the effect of acute exercise on subsequent set shifting performance by exercise intensity, type of exercise, participants' age, and type of control group were examined. Results: Twenty-two studies (N = 1,900) were included into analysis. All aggregated effect sizes ranged from small to moderate. Overall, a small significant beneficial effect was revealed (g = −0.32, 95 % CI −0.45 to −0.18). Heterogeneity of included effect sizes was moderate and significant (T2 = 0.0715, I2 = 46.4%, (p < 0.0016). Moderator analyses revealed a larger average effect in older adults than for studies examining younger adults (−0.42 vs. −0.29). Light exercise (−0.51) led to larger effects than moderate (−0.24) or vigorous exercise (−0.29). Studies testing acute exercise against active control groups showed a noticeably smaller average effect (−0.13) than studies that used passive (−0.38) or cognitive engaging control groups (−0.34). Interestingly, application of resistance or aerobic exercise led to no different average effect sizes (−0.30 vs. −0.32). However, none of the tested covariates reached statistical significance. Conclusion: Acute exercise improves subsequent set shifting performance. However, effect sizes are small, making the relevance for everyday life questionable. The results indicate that older adults benefit more from acute exercise than younger adults do. Light intensity exercise seems most effective while the type of exercise does not seem to influence the magnitude of effects. Research designs with active control groups show the smallest average effect, raising concerns about placebo effects. PROSPERO registration number: CRD42019138799
Collapse
Affiliation(s)
- Max Oberste
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Sophia Sharma
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, Technical University Dortmund, Dortmund, Germany
| |
Collapse
|
9
|
Treble-Barna A, Patronick J, Uchani S, Marousis NC, Zigler CK, Fink EL, Kochanek PM, Conley YP, Yeates KO. Epigenetic Effects on Pediatric Traumatic Brain Injury Recovery (EETR): An Observational, Prospective, Longitudinal Concurrent Cohort Study Protocol. Front Neurol 2020; 11:460. [PMID: 32595586 PMCID: PMC7303323 DOI: 10.3389/fneur.2020.00460] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction: Unexplained heterogeneity in outcomes following pediatric traumatic brain injury (TBI) is one of the most critical barriers to the development of effective prognostic tools and therapeutics. The addition of personal biological factors to our prediction models may account for a significant portion of unexplained variance and advance the field toward precision rehabilitation medicine. The overarching goal of the Epigenetic Effects on Pediatric Traumatic Brain Injury Recovery (EETR) study is to investigate an epigenetic biomarker involved in both childhood adversity and postinjury neuroplasticity to better understand heterogeneity in neurobehavioral outcomes following pediatric TBI. Our primary hypothesis is that childhood adversity will be associated with worse neurobehavioral recovery in part through an epigenetically mediated reduction in brain-derived neurotrophic factor (BDNF) expression in response to TBI. Methods and analysis: EETR is an observational, prospective, longitudinal concurrent cohort study of children aged 3-18 years with either TBI (n = 200) or orthopedic injury (n = 100), recruited from the UPMC Children's Hospital of Pittsburgh. Participants complete study visits acutely and at 6 and 12 months postinjury. Blood and saliva biosamples are collected at all time points-and cerebrospinal fluid (CSF) when available acutely-for epigenetic and proteomic analysis of BDNF. Additional measures assess injury characteristics, pre- and postinjury child neurobehavioral functioning, childhood adversity, and potential covariates/confounders. Recruitment began in July 2017 and will occur for ~6 years, with data collection complete by mid-2023. Analyses will characterize BDNF DNA methylation and protein levels over the recovery period and investigate this novel biomarker as a potential biological mechanism underlying the known association between childhood adversity and worse neurobehavioral outcomes following pediatric TBI. Ethics and dissemination: The study received ethics approval from the University of Pittsburgh Institutional Review Board. Participants and their parents provide informed consent/assent. Research findings will be disseminated via local and international conference presentations and manuscripts submitted to peer-reviewed journals. Trial Registration: The study is registered with clinicaltrials.org (ClinicalTrials.gov Identifier: NCT04186429).
Collapse
Affiliation(s)
- Amery Treble-Barna
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jamie Patronick
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Srivatsan Uchani
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Noelle C. Marousis
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Christina K. Zigler
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, United States
| | - Ericka L. Fink
- Safar Center for Resuscitation Research, Division of Pediatric Critical Care Medicine, UPMC Children's Hospital of Pittsburgh, Department of Critical Care and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, Division of Pediatric Critical Care Medicine, UPMC Children's Hospital of Pittsburgh, Department of Critical Care and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yvette P. Conley
- Department of Health Promotion and Development, School of Nursing, University of Pittsburgh, Pittsburgh, PA, United States
| | - Keith Owen Yeates
- Department of Psychology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
10
|
Marrocco J, Einhorn NR, Petty GH, Li H, Dubey N, Hoffman J, Berman KF, Goldman D, Lee FS, Schmidt PJ, McEwen BS. Epigenetic intersection of BDNF Val66Met genotype with premenstrual dysphoric disorder transcriptome in a cross-species model of estradiol add-back. Mol Psychiatry 2020; 25:572-583. [PMID: 30356121 PMCID: PMC7042769 DOI: 10.1038/s41380-018-0274-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023]
Abstract
Premenstrual dysphoric disorder (PMDD) affects over 5% of women, with symptoms similar to anxiety and major depression, and is associated with differential sensitivity to circulating ovarian hormones. Little is known about the genetic and epigenetic factors that increase the risk to develop PMDD. We report that 17β-estradiol (E2) affects the behavior and the epigenome in a mouse model carrying a single-nucleotide polymorphism of the brain-derived neurotrophic factor gene (BDNF Val66Met), in a way that recapitulates the hallmarks of PMDD. Ovariectomized mice heterozygous for the BDNF Met allele (Het-Met) and their matched wild-type (WT) mice were administered estradiol or vehicle in drinking water for 6 weeks. Using the open field and the splash test, we show that E2 add-back induces anxiety-like and depression-like behavior in Het-Met mice, but not in WT mice. RNA-seq of the ventral hippocampus (vHpc) highlights that E2-dependent gene expression is markedly different between WT mice and Het-Met mice. Through a comparative whole-genome RNA-seq analysis between mouse vHpc and lymphoblastoid cell line cultures from control women and women with PMDD, we discovered common epigenetic biomarkers that transcend species and cell types. Those genes include epigenetic modifiers of the ESC/E(Z) complex, an effector of response to ovarian steroids. Although the BDNF Met genotype intersects the behavioral and transcriptional traits of women with PMDD, we suggest that these similarities speak to the epigenetic factors by which ovarian steroids produce negative behavioral effects.
Collapse
Affiliation(s)
- Jordan Marrocco
- 0000 0001 2166 1519grid.134907.8Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Nathan R. Einhorn
- 0000 0001 2166 1519grid.134907.8Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Gordon H. Petty
- 0000 0001 2166 1519grid.134907.8Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Howard Li
- 0000 0004 0464 0574grid.416868.5Behavioral Endocrinology Branch, National Institute of Mental Health, Bethesda, MD USA
| | - Neelima Dubey
- grid.440681.fDr. D. Y. Patil Biotechnology & Bioinformatics Institute, Pune, India
| | - Jessica Hoffman
- 0000 0001 0421 5525grid.265436.0Uniformed Services University of the Health Sciences, Bethesda, MD USA
| | - Karen F. Berman
- 0000 0004 0464 0574grid.416868.5Section on Integrative Neuroimaging, National Institute of Mental Health, Bethesda, MD USA
| | - David Goldman
- 0000 0004 0481 4802grid.420085.bLaboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD USA
| | - Francis S. Lee
- 000000041936877Xgrid.5386.8Department of Psychiatry, Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, NY USA
| | - Peter J. Schmidt
- 0000 0004 0464 0574grid.416868.5Behavioral Endocrinology Branch, National Institute of Mental Health, Bethesda, MD USA
| | - Bruce S. McEwen
- 0000 0001 2166 1519grid.134907.8Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| |
Collapse
|
11
|
Costa CM, Oliveira GLD, Fonseca ACS, Lana RDC, Polese JC, Pernambuco AP. Levels of cortisol and neurotrophic factor brain-derived in Parkinson’s disease. Neurosci Lett 2019; 708:134359. [DOI: 10.1016/j.neulet.2019.134359] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 01/01/2023]
|
12
|
Wang Q, Liu J, Guo Y, Dong G, Zou W, Chen Z. Association between BDNF G196A (Val66Met) polymorphism and cognitive impairment in patients with Parkinson's disease: a meta-analysis. ACTA ACUST UNITED AC 2019; 52:e8443. [PMID: 31365694 PMCID: PMC6668960 DOI: 10.1590/1414-431x20198443] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 05/17/2019] [Indexed: 02/08/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is widely expressed in the central nervous system and prolongs the survival of dopaminergic neurons in the substantia nigra. Several studies have recently investigated the association between BDNF G196A (Val66Met), a single nucleotide polymorphism influencing cognitive processes, and cognitive impairment in Parkinson's disease (PD), but with contradictory findings. Thus, this meta-analysis was performed to clarify the possible association. Relevant studies were identified by a systematic search of PubMed, Embase, and China National Knowledge Infrastructure (CNKI) databases. The strength of the association was evaluated using crude odds ratios and 95% confidence interval. Finally, six studies involving 532 cases and 802 controls were included. Our analyses suggested the G196A (Val66Met) polymorphism was significantly associated with cognitive impairment in PD, especially in Caucasian populations. In conclusion, BDNF G196A (Val66Met) is confirmed to be a risk factor for cognitive impairment in PD.
Collapse
Affiliation(s)
- Qian Wang
- Department of Neurology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Jianfang Liu
- Department of Neurology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yikun Guo
- Department of Neurology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Guanzhong Dong
- Department of Neurology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Wenying Zou
- Department of Neurology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Zhuoyou Chen
- Department of Neurology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
13
|
Harper SA, Dowdell BT, Kim JH, Pollock BS, Ridgel AL. Non-Motor Symptoms after One Week of High Cadence Cycling in Parkinson's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E2104. [PMID: 31197095 PMCID: PMC6616554 DOI: 10.3390/ijerph16122104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/23/2022]
Abstract
The objective was to investigate if high cadence cycling altered non-motor cognition and depression symptoms in individuals with Parkinson's disease (PD) and whether exercise responses were influenced by brain-derived neurotrophic factor (BDNF) Val66Met polymorphism. Individuals with idiopathic PD who were ≥50 years old and free of surgical procedures for PD were recruited. Participants were assigned to either a cycling (n = 20) or control (n = 15) group. The cycling group completed three sessions of high cadence cycling on a custom motorized stationary ergometer. The primary outcome was cognition (attention, executive function, and emotion recognition were assessed via WebNeuro® and global cognition via Montreal Cognitive Assessment). Depression symptoms were assessed via Beck Depression Inventory-II. There was a main effect of time for emotional recognition (p = 0.048), but there were no other changes in cognition or depression symptoms. Regardless of intervention or Val66Met polymorphism, high cadence cycling does not alter cognition or depression symptoms after three sessions in one week.
Collapse
Affiliation(s)
- Sara A Harper
- Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA.
- Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL 35205, USA.
| | - Bryan T Dowdell
- Exercise Physiology Department, Kent State University, Kent, OH 44240, USA.
| | - Jin Hyun Kim
- Exercise Physiology Department, Kent State University, Kent, OH 44240, USA.
| | - Brandon S Pollock
- Department of Exercise Science, Walsh University, North Canton, OH 44720, USA.
| | - Angela L Ridgel
- Exercise Physiology Department, Kent State University, Kent, OH 44240, USA.
| |
Collapse
|
14
|
BDNF Val66Met polymorphism and cognitive impairment in Parkinson's disease-a meta-analysis. Neurol Sci 2019; 40:1901-1907. [PMID: 31104169 DOI: 10.1007/s10072-019-03907-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 04/16/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Conflicting results identifying the association between Brain-derived neurotrophic factor (BDNF) polymorphism, Val66Met, and cognitive impairment in Parkinson's disease (PD) have been reported. METHODS AND RESULTS To clarify whether Val66Met is related to cognitive impairment in PD, we carried out this meta-analysis by searching literature from PubMed, Web of Science, and Embase databases regarding this polymorphism. Six eligible studies involving 1467 PD patients were included in this meta-analysis. Our results showed statistically significant association between Val66Met and risk of cognitive impairment in PD patients in additive model (Met/Met vs. Val/Val: OR 3.82, 95%CI 1.32 to 11.08, p = 0.01) and recessive model (Met/Met vs. Val-carrier: OR 3.81, 95%CI 1.38 to 10.53, p = 0.01) except for dominant model. CONCLUSIONS Our meta-analysis implicates Val66Met BDNF polymorphism may be associated with Parkinson's disease cognitive impairment, further well-designed studies with larger populations are required to validate these results owing to the limited research.
Collapse
|
15
|
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: 2.5] [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
|
16
|
Sun MK. Executive functioning: perspectives on neurotrophic activity and pharmacology. Behav Pharmacol 2018; 29:592-604. [PMID: 30179884 DOI: 10.1097/fbp.0000000000000427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Executive functioning is a high-level cognitive ability, regulating other abilities and behaviors to achieve desired goals. A typical executive task can be defined as the capacity to maintain one's attention on the current task, that is, responding only to the correct but not to distractive stimuli. Impairments of executive functions, or executive dysfunctions, have a growing impact on everyday life and academic achievement and are usually an early feature, and one of the core features, in brain injury and memory and behavioral disorders. Furthermore, emerging evidence indicates that memory therapeutics cannot achieve their clinical benefits in cognition if executive dysfunction is not effectively and simultaneously treated. Improvement of executive functions might be achieved through targeting some signaling pathways in the brain, including the brain-derived neurotrophic factor signaling pathways. These agents may be useful either as stand-alone interventions for patients with executive dysfunction and/or psychiatric and memory disorders or as essential adjuncts to drugs that target the underlying pathology in various brain injury and memory and behavioral disorders.
Collapse
Affiliation(s)
- Miao-Kun Sun
- Blanchette Rockefeller Neurosciences Institute, Morgantown, West Virginia, USA
| |
Collapse
|
17
|
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: 66] [Impact Index Per Article: 11.0] [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
|
18
|
Shen T, You Y, Joseph C, Mirzaei M, Klistorner A, Graham SL, Gupta V. BDNF Polymorphism: A Review of Its Diagnostic and Clinical Relevance in Neurodegenerative Disorders. Aging Dis 2018; 9:523-536. [PMID: 29896439 PMCID: PMC5988606 DOI: 10.14336/ad.2017.0717] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/17/2017] [Indexed: 12/17/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has a unique role in the neuronal development, differentiation, and survival in the developing and adult nervous system. A common single-nucleotide polymorphism in the pro-region of the human BDNF gene, resulting in a valine to methionine substitution (Val66Met), has been associated with the susceptibility, incidence, and clinical features of several neurodegenerative disorders. Much research has been dedicated to evaluating the effects of polymorphism in the past decade, and functional effects of this genetic variation. A better understanding of how this naturally occurring polymorphism associates with or influences physiology, anatomy, and cognition in both healthy and diseased adults in neurodegenerative conditions will help understand neurochemical mechanisms and definable clinical outcomes in humans. Here we review the role and relevance of the BDNF Val66Met polymorphism in neurodegenerative diseases, with particular emphasis on glaucoma, multiple sclerosis (MS), Alzheimer’s disease (AD) and Parkinson’s disease (PD). Several controversies and unresolved issues, including small effect sizes, possible ethnicity, gender, and age effects of the BDNF Val66Met are also discussed with respect to future research.
Collapse
Affiliation(s)
- Ting Shen
- 1Faculty of Medicine and Health Sciences, Macquarie University, Australia
| | - Yuyi You
- 2Save Sight Institute, Sydney University, Sydney, Australia
| | - Chitra Joseph
- 1Faculty of Medicine and Health Sciences, Macquarie University, Australia
| | - Mehdi Mirzaei
- 3Faculty of Science and Engineering, Macquarie University, Australia
| | - Alexander Klistorner
- 1Faculty of Medicine and Health Sciences, Macquarie University, Australia.,2Save Sight Institute, Sydney University, Sydney, Australia
| | - Stuart L Graham
- 1Faculty of Medicine and Health Sciences, Macquarie University, Australia.,2Save Sight Institute, Sydney University, Sydney, Australia
| | - Vivek Gupta
- 1Faculty of Medicine and Health Sciences, Macquarie University, Australia
| |
Collapse
|
19
|
Li J, Zhang S, Li C, Li M, Ma L. Sitagliptin rescues memory deficits in Parkinsonian rats via upregulating BDNF to prevent neuron and dendritic spine loss. Neurol Res 2018; 40:736-743. [PMID: 29781786 DOI: 10.1080/01616412.2018.1474840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Parkinson's disease (PD) is a neurodegenerative disease with high morbidity among adults worldwide that causes tremendous trouble to people's lives. The purpose of this study was to investigate the impact of sitagliptin on PD and its potential mechanism. METHODS First, the memory of rats in each group was evaluated with the Morris water maze (MWM) test and the passive avoidance test. Then, both brain-derived neurotrophic factor (BDNF) protein and mRNA levels were detected by ELISA and qPCR assays, respectively. Then, rapid Golgi impregnation was used to observe the density of dendritic spines in the hippocampal CA1 area. Finally, k252a, an antagonist of Trk receptors, was used to block the binding of BDNF with its receptors, and the effects of sitagliptin on PD improvement were detected. RESULTS Our study showed that sitagliptin improved memory deficits in PD rats. Meanwhile, the expression level of BDNF and tyrosine hydroxylase (TH) was upregulated, and the density of dendritic spine was increased by sitagliptin administration. Moreover, K252a administration blocked the positive effects of sitagliptin on memory in PD rats. DISCUSSION Sitagliptin rescued the memory deficits, which was achieved by upregulating BDNF to prevent neuronal death and dendritic spine loss. Our findings indicate that sitagliptin might be a promising potential drug for PD treatment in the future.
Collapse
Affiliation(s)
- Jing Li
- a Department of Geriatrics , the Second Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang P.R. China
| | - Shuhu Zhang
- a Department of Geriatrics , the Second Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang P.R. China
| | - Chenye Li
- a Department of Geriatrics , the Second Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang P.R. China
| | - Mei Li
- a Department of Geriatrics , the Second Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang P.R. China
| | - Lan Ma
- a Department of Geriatrics , the Second Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang P.R. China
| |
Collapse
|
20
|
Hirsch MA, van Wegen EEH, Newman MA, Heyn PC. Exercise-induced increase in brain-derived neurotrophic factor in human Parkinson's disease: a systematic review and meta-analysis. Transl Neurodegener 2018; 7:7. [PMID: 29568518 PMCID: PMC5859548 DOI: 10.1186/s40035-018-0112-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/28/2018] [Indexed: 01/02/2023] Open
Abstract
Background Animal models of exercise and Parkinson’s disease (PD) have found that the physiologic use of exercise may interact with the neurodegenerative disease process, likely mediated by brain derived neurotrophic factor (BDNF). No reviews so far have assessed the methodologic quality of available intervention studies or have bundled the effect sizes of individual studies on exercise-induced effects on BDNF blood levels in human PD. Research design and methods We searched MEDLINE, EMBASE, Cochrane Library, PsycINFO and PubMed from inception to June 2017. Results Data aggregated from two randomized controlled trials and four pre-experimental studies with a total of 100 ambulatory patients with idiopathic PD (Hoehn/Yahr ≤3) found improvements in BDNF blood concentration levels in all 6 studies (two RCTs and 4 pre-experimental studies). Pooled BDNF level change scores from the 2 RCTs resulted in a significant homogeneous summary effect size (Standardized Mean Difference 2.06, 95% CI 1.36 to 2.76), and a significant heterogeneous SES for the motor part of the UPDRS-III examination (MD -5.53, 95% CI -10.42 to -0.64). Clinical improvements were noted in all studies using a variety of outcome measures. Limitations The evidence-base consists primarily of small studies with low to moderate methodological quality. Conclusions This review provides preliminary evidence for the effectiveness of physical exercise treatments for persons with PD on BDNF blood levels. Further research is needed. Electronic supplementary material The online version of this article (10.1186/s40035-018-0112-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mark A Hirsch
- 1Carolinas Medical Center, Carolinas Rehabilitation, Department of Physical Medicine and Rehabilitation, 1100 Blythe Blvd, Charlotte, NC 28203 USA
| | - Erwin E H van Wegen
- 2Department of Rehabilitation Medicine, Amsterdam Movement Sciences/Amsterdam Neurosciences, VU University Medical Center, PO Box 7057, 1007 Amsterdam, MB The Netherlands
| | - Mark A Newman
- 1Carolinas Medical Center, Carolinas Rehabilitation, Department of Physical Medicine and Rehabilitation, 1100 Blythe Blvd, Charlotte, NC 28203 USA
| | - Patricia C Heyn
- 3Department of Physical Medicine and Rehabilitation, Anschutz Medical Campus, University of Colorado, Denver, USA
| |
Collapse
|
21
|
Almeida MF, Silva CM, Chaves RS, Lima NCR, Almeida RS, Melo KP, Demasi M, Fernandes T, Oliveira EM, Netto LES, Cardoso SM, Ferrari MFR. Effects of mild running on substantia nigra during early neurodegeneration. J Sports Sci 2017; 36:1363-1370. [PMID: 28895489 DOI: 10.1080/02640414.2017.1378494] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Moderate physical exercise acts at molecular and behavioural levels, such as interfering in neuroplasticity, cell death, neurogenesis, cognition and motor functions. Therefore, the aim of this study is to analyse the cellular effects of moderate treadmill running upon substantia nigra during early neurodegeneration. Aged male Lewis rats (9-month-old) were exposed to rotenone 1mg/kg/day (8 weeks) and 6 weeks of moderate treadmill running, beginning 4 weeks after rotenone exposure. Substantia nigra was extracted and submitted to proteasome and antioxidant enzymes activities, hydrogen peroxide levels and Western blot to evaluate tyrosine hydroxylase (TH), alpha-synuclein, Tom-20, PINK1, TrkB, SLP1, CRMP-2, Rab-27b, LC3II and Beclin-1 level. It was demonstrated that moderate treadmill running, practiced during early neurodegeneration, prevented the increase of alpha-synuclein and maintained the levels of TH unaltered in substantia nigra of aged rats. Physical exercise also stimulated autophagy and prevented impairment of mitophagy, but decreased proteasome activity in rotenone-exposed aged rats. Physical activity also prevented H2O2 increase during early neurodegeneration, although the involved mechanism remains to be elucidated. TrkB levels and its anterograde trafficking seem not to be influenced by moderate treadmill running. In conclusion, moderate physical training could prevent early neurodegeneration in substantia nigra through the improvement of autophagy and mitophagy.
Collapse
Affiliation(s)
- Michael F Almeida
- a Departamento de Genética e Biologia Evolutiva, Instituto de Biociências , Universidade de São Paulo , São Paulo , Brazil
| | - Carolliny M Silva
- a Departamento de Genética e Biologia Evolutiva, Instituto de Biociências , Universidade de São Paulo , São Paulo , Brazil
| | - Rodrigo S Chaves
- a Departamento de Genética e Biologia Evolutiva, Instituto de Biociências , Universidade de São Paulo , São Paulo , Brazil
| | - Nathan C R Lima
- a Departamento de Genética e Biologia Evolutiva, Instituto de Biociências , Universidade de São Paulo , São Paulo , Brazil
| | - Renato S Almeida
- b Institute for Biosciences , University of Taubate , Taubate , Brazil
| | - Karla P Melo
- a Departamento de Genética e Biologia Evolutiva, Instituto de Biociências , Universidade de São Paulo , São Paulo , Brazil
| | - Marilene Demasi
- c Laboratory of Biochemistry and Biophysics , Butantan Institute , Sao Paulo , Brazil
| | - Tiago Fernandes
- d Laboratory of Biochemistry and Molecular Biology of the Exercise, Department of Human Movement Biodynamic, School of Physical Education and Sport , University of Sao Paulo , Sao Paulo , Brazil
| | - Edilamar M Oliveira
- d Laboratory of Biochemistry and Molecular Biology of the Exercise, Department of Human Movement Biodynamic, School of Physical Education and Sport , University of Sao Paulo , Sao Paulo , Brazil
| | - Luis E S Netto
- a Departamento de Genética e Biologia Evolutiva, Instituto de Biociências , Universidade de São Paulo , São Paulo , Brazil
| | - Sandra M Cardoso
- e Center for Neuroscience and Cell Biology , University of Coimbra , Coimbra , Portugal.,f Institute of Cellular and Molecular Biology, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Merari F R Ferrari
- a Departamento de Genética e Biologia Evolutiva, Instituto de Biociências , Universidade de São Paulo , São Paulo , Brazil
| |
Collapse
|
22
|
Caspell-Garcia C, Simuni T, Tosun-Turgut D, Wu IW, Zhang Y, Nalls M, Singleton A, Shaw LA, Kang JH, Trojanowski JQ, Siderowf A, Coffey C, Lasch S, Aarsland D, Burn D, Chahine LM, Espay AJ, Foster ED, Hawkins KA, Litvan I, Richard I, Weintraub D. Multiple modality biomarker prediction of cognitive impairment in prospectively followed de novo Parkinson disease. PLoS One 2017; 12:e0175674. [PMID: 28520803 PMCID: PMC5435130 DOI: 10.1371/journal.pone.0175674] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 03/29/2017] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES To assess the neurobiological substrate of initial cognitive decline in Parkinson's disease (PD) to inform patient management, clinical trial design, and development of treatments. METHODS We longitudinally assessed, up to 3 years, 423 newly diagnosed patients with idiopathic PD, untreated at baseline, from 33 international movement disorder centers. Study outcomes were four determinations of cognitive impairment or decline, and biomarker predictors were baseline dopamine transporter (DAT) single photon emission computed tomography (SPECT) scan, structural magnetic resonance imaging (MRI; volume and thickness), diffusion tensor imaging (mean diffusivity and fractional anisotropy), cerebrospinal fluid (CSF; amyloid beta [Aβ], tau and alpha synuclein), and 11 single nucleotide polymorphisms (SNPs) previously associated with PD cognition. Additionally, longitudinal structural MRI and DAT scan data were included. Univariate analyses were run initially, with false discovery rate = 0.2, to select biomarker variables for inclusion in multivariable longitudinal mixed-effect models. RESULTS By year 3, cognitive impairment was diagnosed in 15-38% participants depending on the criteria applied. Biomarkers, some longitudinal, predicting cognitive impairment in multivariable models were: (1) dopamine deficiency (decreased caudate and putamen DAT availability); (2) diffuse, cortical decreased brain volume or thickness (frontal, temporal, parietal, and occipital lobe regions); (3) co-morbid Alzheimer's disease Aβ amyloid pathology (lower CSF Aβ 1-42); and (4) genes (COMT val/val and BDNF val/val genotypes). CONCLUSIONS Cognitive impairment in PD increases in frequency 50-200% in the first several years of disease, and is independently predicted by biomarker changes related to nigrostriatal or cortical dopaminergic deficits, global atrophy due to possible widespread effects of neurodegenerative disease, co-morbid Alzheimer's disease plaque pathology, and genetic factors.
Collapse
Affiliation(s)
- Chelsea Caspell-Garcia
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, United States of America
| | - Tanya Simuni
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Duygu Tosun-Turgut
- University of California, San Francisco, San Francisco, CA, United States of America
| | - I-Wei Wu
- University of California, San Francisco, San Francisco, CA, United States of America
| | - Yu Zhang
- University of California, San Francisco, San Francisco, CA, United States of America
| | - Mike Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
| | - Leslie A. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States of America
| | - Ju-Hee Kang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Pharmacology & Clinical Pharmacology, Inha University School of Medicine, Incheon, Republic of Korea
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States of America
| | - Andrew Siderowf
- Avid Radiopharmaceuticals, Philadelphia, PA, United States of America
| | - Christopher Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, United States of America
| | - Shirley Lasch
- Institute for Neurodegenerative Disorders (IND) and Molecular NeuroImaging, LLC (MNI), New Haven CT, United States of America
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, England
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - David Burn
- Institute for Ageing and Health, Newcastle University, Newcastle, England
| | - Lana M. Chahine
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States of America
| | - Alberto J. Espay
- Department of Neurology, University of Cincinnati Academic Health Center, Cincinnati, OH, United States of America
| | - Eric D. Foster
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, United States of America
| | - Keith A. Hawkins
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States of America
| | - Irene Litvan
- UCSD Movement Disorder Center, Department of Neurosciences, University of California San Diego, San Diego, CA, United States of America
| | - Irene Richard
- Departments of Neurology and Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Daniel Weintraub
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States of America
- Parkinson’s Disease Research, Education and Clinical Center (PADRECC and MIRECC), Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, United States of America
- Mental Illness Research, Education and Clinical Center (MIRECC), Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, United States of America
| | | |
Collapse
|
23
|
Fagan ES, Pihlstrøm L. Genetic risk factors for cognitive decline in Parkinson's disease: a review of the literature. Eur J Neurol 2017; 24:561-e20. [DOI: 10.1111/ene.13258] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/11/2017] [Indexed: 01/18/2023]
Affiliation(s)
- E. S. Fagan
- Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - L. Pihlstrøm
- Institute of Clinical Medicine; University of Oslo; Oslo Norway
- Department of Neurology; Oslo University Hospital; Oslo Norway
| |
Collapse
|
24
|
Asakawa T, Fang H, Sugiyama K, Nozaki T, Kobayashi S, Hong Z, Suzuki K, Mori N, Yang Y, Hua F, Ding G, Wen G, Namba H, Xia Y. Human behavioral assessments in current research of Parkinson's disease. Neurosci Biobehav Rev 2016; 68:741-772. [PMID: 27375277 DOI: 10.1016/j.neubiorev.2016.06.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is traditionally classified as a movement disorder because patients mainly complain about motor symptoms. Recently, non-motor symptoms of PD have been recognized by clinicians and scientists as early signs of PD, and they are detrimental factors in the quality of life in advanced PD patients. It is crucial to comprehensively understand the essence of behavioral assessments, from the simplest measurement of certain symptoms to complex neuropsychological tasks. We have recently reviewed behavioral assessments in PD research with animal models (Asakawa et al., 2016). As a companion volume, this article will systematically review the behavioral assessments of motor and non-motor PD symptoms of human patients in current research. The major aims of this article are: (1) promoting a comparative understanding of various behavioral assessments in terms of the principle and measuring indexes; (2) addressing the major strengths and weaknesses of these behavioral assessments for a better selection of tasks/tests in order to avoid biased conclusions due to inappropriate assessments; and (3) presenting new concepts regarding the development of wearable devices and mobile internet in future assessments. In conclusion we emphasize the importance of improving the assessments for non-motor symptoms because of their complex and unique mechanisms in human PD brains.
Collapse
Affiliation(s)
- Tetsuya Asakawa
- Department of Neurosurgery, Hamamatsu University School of Medicine, Handayama, Hamamatsu-city, Shizuoka, Japan; Department of Psychiatry, Hamamatsu University School of Medicine, Handayama, Hamamatsu-city, Shizuoka, Japan.
| | - Huan Fang
- Department of Pharmacy, Jinshan Hospital of Fudan University, Shanghai, China
| | - Kenji Sugiyama
- Department of Neurosurgery, Hamamatsu University School of Medicine, Handayama, Hamamatsu-city, Shizuoka, Japan
| | - Takao Nozaki
- Department of Neurosurgery, Hamamatsu University School of Medicine, Handayama, Hamamatsu-city, Shizuoka, Japan
| | - Susumu Kobayashi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Handayama, Hamamatsu-city, Shizuoka, Japan
| | - Zhen Hong
- Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China
| | - Katsuaki Suzuki
- Department of Psychiatry, Hamamatsu University School of Medicine, Handayama, Hamamatsu-city, Shizuoka, Japan
| | - Norio Mori
- Department of Psychiatry, Hamamatsu University School of Medicine, Handayama, Hamamatsu-city, Shizuoka, Japan
| | - Yilin Yang
- The First People's Hospital of Changzhou, Soochow University School of Medicine, Changzhou, China
| | - Fei Hua
- The First People's Hospital of Changzhou, Soochow University School of Medicine, Changzhou, China
| | - Guanghong Ding
- Shanghai Key laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Guoqiang Wen
- Department of Neurology, Hainan General Hospital, Haikou, Hainan, China
| | - Hiroki Namba
- Department of Neurosurgery, Hamamatsu University School of Medicine, Handayama, Hamamatsu-city, Shizuoka, Japan
| | - Ying Xia
- Department of Neurosurgery, The University of Texas McGovern Medical School, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Kim HJ, Jeon B. How close are we to individualized medicine for Parkinson's disease? Expert Rev Neurother 2016; 16:815-30. [PMID: 27105072 DOI: 10.1080/14737175.2016.1182021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION There is a considerable inter-individual heterogeneity in clinical features, disease course, and treatment response in Parkinson's disease (PD), which can be explained not only by disease process and clinical variables, but also by an impact from genetic factors. Evidence-based medicine relies on large randomized control trials and meta-analysis-average medicine, which ignores individual differences. However, we are now in the early phases of a paradigm shift in medicine relating to individuality and variability. The purpose of individualized medicine is to predict patients' responses to targeted therapy using diagnostic tests based on genetics or other molecular mechanisms, thus providing the right drug at the right dose at the right time. AREAS COVERED In this article, we outline current state of individualized medicine for PD. Expert Commentary: Pharmacogenomics, an important element of individualized medicine, is just beginning to be considered in PD. To advance the clinical use of pharmacogenomics, big data cohort for genomic research and multidisciplinary team approaches are necessary.
Collapse
Affiliation(s)
- Hee Jin Kim
- a Department of Neurology , Konkuk University Medical Center , Seoul , South Korea.,b Parkinson Disease Study Group , Seoul National University Hospital , Seoul , South Korea
| | - Beomseok Jeon
- a Department of Neurology , Konkuk University Medical Center , Seoul , South Korea.,c Department of Neurology and Movement Disorder Center, College of Medicine , Seoul National University , Seoul , South Korea
| |
Collapse
|
26
|
Aliperti V, Donizetti A. Long Non-coding RNA in Neurons: New Players in Early Response to BDNF Stimulation. Front Mol Neurosci 2016; 9:15. [PMID: 26973456 PMCID: PMC4773593 DOI: 10.3389/fnmol.2016.00015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/18/2016] [Indexed: 12/28/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin family member that is highly expressed and widely distributed in the brain. BDNF is critical for neural survival and plasticity both during development and in adulthood, and dysfunction in its signaling may contribute to a number of neurodegenerative disorders. Deep understanding of the BDNF-activated molecular cascade may thus help to find new biomarkers and therapeutic targets. One interesting direction is related to the early phase of BDNF-dependent gene expression regulation, which is responsible for the activation of selective gene programs that lead to stable functional and structural remodeling of neurons. Immediate-early coding genes activated by BDNF are under investigation, but the involvement of the non-coding RNAs is largely unexplored, especially the long non-coding RNAs (lncRNAs). lncRNAs are emerging as key regulators that can orchestrate different aspects of nervous system development, homeostasis, and plasticity, making them attractive candidate markers and therapeutic targets for brain diseases. We used microarray technology to identify differentially expressed lncRNAs in the immediate response phase of BDNF stimulation in a neuronal cell model. Our observations on the putative functional role of lncRNAs provide clues to their involvement as master regulators of gene expression cascade triggered by BDNF.
Collapse
Affiliation(s)
- Vincenza Aliperti
- Department of Biology, University of Naples Federico II Naples, Italy
| | - Aldo Donizetti
- Department of Biology, University of Naples Federico II Naples, Italy
| |
Collapse
|
27
|
Shpektor A, Bartrés-Faz D, Feurra M. Commentary: Duration-dependent effects of the BDNF Val66Met polymorphism on anodal tDCS induced motor cortex plasticity in older adults: a group and individual perspective. Front Aging Neurosci 2015; 7:183. [PMID: 26441642 PMCID: PMC4585066 DOI: 10.3389/fnagi.2015.00183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/07/2015] [Indexed: 01/24/2023] Open
Affiliation(s)
- Anna Shpektor
- School of Psychology, Centre for Cognition and Decision Making, National Research University Higher School of Economics Moscow, Russia
| | - David Bartrés-Faz
- Department of Psychiatry and Clinical Psychobiology, University of Barcelona Barcelona, Spain
| | - Matteo Feurra
- School of Psychology, Centre for Cognition and Decision Making, National Research University Higher School of Economics Moscow, Russia ; Unit of Neurology and Clinical Neurophysiology, Brain Investigation and Neuromodulation laboratory (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Azienda Ospedaliera Universitaria of Siena Siena, Italy
| |
Collapse
|
28
|
Ng T, Teo SM, Yeo HL, Shwe M, Gan YX, Cheung YT, Foo KM, Cham MT, Lee JA, Tan YP, Fan G, Yong WS, Preetha M, Loh WJK, Koo SL, Jain A, Lee GE, Wong M, Dent R, Yap YS, Ng R, Khor CC, Ho HK, Chan A. Brain-derived neurotrophic factor genetic polymorphism (rs6265) is protective against chemotherapy-associated cognitive impairment in patients with early-stage breast cancer. Neuro Oncol 2015; 18:244-51. [PMID: 26289590 PMCID: PMC4724179 DOI: 10.1093/neuonc/nov162] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/17/2015] [Indexed: 12/31/2022] Open
Abstract
Background Brain-derived neurotrophic factor (BDNF), a neurotrophin that regulates neuronal function and development, is implicated in several neurodegenerative conditions. Preliminary data suggest that a reduction of BDNF concentrations may lead to postchemotherapy cognitive impairment. We hypothesized that a single nucleotide polymorphism (rs6265) of the BDNF gene may predispose patients to cognitive impairment. This study aimed to evaluate the effect of BDNF gene polymorphism on chemotherapy-associated cognitive impairment. Methods Overall, 145 patients receiving chemotherapy for early-stage breast cancer (mean age: 50.8 ± 8.8 y; 82.1% Chinese) were recruited. Patients' cognitive functions were assessed longitudinally using the validated Functional Assessment of Cancer Therapy–Cognitive Function (v.3) and an objective computerized tool, Headminder. Genotyping was performed using Sanger sequencing. Logistic regression was used to evaluate the association between BDNF Val66Met polymorphism and cognition after adjusting for ethnicity and clinically important covariates. Results Of the 145 patients, 54 (37%) reported cognitive impairment postchemotherapy. The Met/Met genotype was associated with statistically significant lower odds of developing cognitive impairment (odds ratio [OR] = 0.26; 95% CI: 0.08–0.92; P = .036). The Met carriers were less likely to experience impairment in the domains of verbal fluency (OR = 0.34; 95% CI: 0.12–0.90; P = .031) and multitasking ability (OR = 0.37; 95% CI: 0.15–0.91; P = .030) compared with the Val/Val homozygote. No associations were observed between Headminder and the BDNF Val66Met polymorphism. Conclusions This is the first study to provide evidence that carriers of the BDNF Met allele are protected against chemotherapy-associated cognitive impairment. Further studies are required to validate the findings.
Collapse
Affiliation(s)
- Terence Ng
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Shu Mei Teo
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Hui Ling Yeo
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Maung Shwe
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Yan Xiang Gan
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Yin Ting Cheung
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Koon Mian Foo
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Mooi Tai Cham
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Jung Ah Lee
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Yee Pin Tan
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Gilbert Fan
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Wei Sean Yong
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Madhukumar Preetha
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Wei-Jen Kiley Loh
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Si-Lin Koo
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Amit Jain
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Guek Eng Lee
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Mabel Wong
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Rebecca Dent
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Yoon Sim Yap
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Raymond Ng
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Chiea Chuen Khor
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Han Kiat Ho
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| | - Alexandre Chan
- Department of Pharmacy, National University of Singapore, Singapore (T.N., S.M.T., H.L.Y., M.S., Y.T.C., H.K.H., A.C.); Department of Pharmacy, National Cancer Centre Singapore, Singapore (Y.X.G., A.C.); Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore (K.M.F.); Breast Centre, K.K. Women's and Children's Hospital, Singapore (M.T.C., J.A.L.); Department of Psychosocial Oncology, National Cancer Centre Singapore, Singapore (Y.P.T., G.F.); Department of Surgical Oncology, National Cancer Centre Singapore, Singapore (W.S.Y., M.P.); Department of Medical Oncology, National Cancer Centre Singapore, Singapore (W.-J.K.L., S.-L.K., A.J., G.E.L., M.W., R.D., Y.S.Y., R.N.); Duke-N.U.S Graduate Medical School Singapore, Singapore (R.D., R.N.); Human Genetics, Genome Institute of Singapore, Singapore (C.C.K.); Singapore Eye Research Institute, Singapore (C.C.K.)
| |
Collapse
|
29
|
Du X, Pang TY. Is Dysregulation of the HPA-Axis a Core Pathophysiology Mediating Co-Morbid Depression in Neurodegenerative Diseases? Front Psychiatry 2015; 6:32. [PMID: 25806005 PMCID: PMC4353372 DOI: 10.3389/fpsyt.2015.00032] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/16/2015] [Indexed: 01/19/2023] Open
Abstract
There is increasing evidence of prodromal manifestation of neuropsychiatric symptoms in a variety of neurodegenerative diseases such as Parkinson's disease (PD) and Huntington's disease (HD). These affective symptoms may be observed many years before the core diagnostic symptoms of the neurological condition. It is becoming more apparent that depression is a significant modifying factor of the trajectory of disease progression and even treatment outcomes. It is therefore crucial that we understand the potential pathophysiologies related to the primary condition, which could contribute to the development of depression. The hypothalamic-pituitary-adrenal (HPA)-axis is a key neuroendocrine signaling system involved in physiological homeostasis and stress response. Disturbances of this system lead to severe hormonal imbalances, and the majority of such patients also present with behavioral deficits and/or mood disorders. Dysregulation of the HPA-axis is also strongly implicated in the pathology of major depressive disorder. Consistent with this, antidepressant drugs, such as the selective serotonin reuptake inhibitors have been shown to alter HPA-axis activity. In this review, we will summarize the current state of knowledge regarding HPA-axis pathology in Alzheimer's, PD and HD, differentiating between prodromal and later stages of disease progression when evidence is available. Both clinical and preclinical evidence will be examined, but we highlight animal model studies as being particularly useful for uncovering novel mechanisms of pathology related to co-morbid mood disorders. Finally, we purpose utilizing the preclinical evidence to better inform prospective, intervention studies.
Collapse
Affiliation(s)
- Xin Du
- Mental Health Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne , Melbourne, VIC , Australia
| | - Terence Y Pang
- Behavioural Neurosciences Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne , Melbourne, VIC , Australia
| |
Collapse
|