1
|
Gellé T, Vinais T, Lacroix A, Plansont B, Nubukpo P, Girard M. Serum BDNF and pro-BDNF levels in alcohol use disorders according to depression status: An exploratory study of their evolution two months after withdrawal. Heliyon 2024; 10:e38940. [PMID: 39430530 PMCID: PMC11490827 DOI: 10.1016/j.heliyon.2024.e38940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Background Alcohol use disorders (AUDs) are complex pathologies with a myriad of molecular actors involved in both disease progression and remission. Brain-derived neurotrophic factor (BDNF) is suspected to be one such actor due to its neurotrophic effects. The BDNF precursor, pro-BDNF, has different effects, as it mainly promotes neuronal apoptosis. Both forms also play a role in depression and depressive episodes (DE). The aim of this exploratory study was to compare serum BDNF and pro-BDNF levels in patients with AUDs after withdrawal and according to DE status with those of controls without AUDs or DE. Materials and methods Ninety-nine AUD patients and 40 controls were included. Questionnaires were used to assess both alcohol and psychiatric domains: the severity of hazardous alcohol consumption was assessed using Alcohol Use Disorders Identification Test (AUDIT), craving was assessed using Obsessive and Compulsive Drinking Scale (OCDS), anxiety was assessed with Hamilton Anxiety Rating Scale (HAM-A) and depression with Montgomery-Åsberg Depression Rating Scale (MADRS). Blood samples were collected during two visits: at the time of alcohol withdrawal (M0) and two months later (M2). ELISAs to measure serum BDNF and pro-BDNF levels were performed. AUD patients were categorized according to depression status at M2. Forty-five patients remained abstinent whereas 54 relapsed. BDNF serum levels rose after alcohol withdrawal, but pro-BDNF levels did not vary between M0 and M2. Results AUD subjects without DE at M2 had higher BDNF levels at both M0 and M2 than AUD subjects with DE at M2. AUD subjects showed lower MADRS and OCD scores at M2 than at M0. AUD subjects without DE had lower BDNF levels at M0 than controls but not at M2, regardless of abstinence maintenance. Conclusion BDNF serum levels were reduced in AUD patients compared to controls and were further reduced in patients with both AUDs and DE. Alcohol withdrawal treatment was sufficient to induce an increase in serum BDNF levels after 2 months, regardless of whether abstinence was maintained during this time period.
Collapse
Affiliation(s)
- Thibaut Gellé
- Inserm U1094, IRD UMR270, Univ. Limoges, CHU Limoges, EpiMaCT - Epidemiology of chronic diseases in tropical zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
| | - Théodore Vinais
- Inserm U1094, IRD UMR270, Univ. Limoges, CHU Limoges, EpiMaCT - Epidemiology of chronic diseases in tropical zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| | - Aurélie Lacroix
- Inserm U1094, IRD UMR270, Univ. Limoges, CHU Limoges, EpiMaCT - Epidemiology of chronic diseases in tropical zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| | - Brigitte Plansont
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| | - Philippe Nubukpo
- Inserm U1094, IRD UMR270, Univ. Limoges, CHU Limoges, EpiMaCT - Epidemiology of chronic diseases in tropical zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| | - Murielle Girard
- Research and Innovation Unit, Esquirol Hospital, 87025, Limoges, France
| |
Collapse
|
2
|
Correia AS, Torrado M, Costa-Coelho T, Carvalho ED, Inteiro-Oliveira S, Diógenes MJ, Pêgo AP, Santos SD, Sebastião AM, Vale N. Brain-derived neurotrophic factor modulation in response to oxidative stress and corticosterone: role of scopolamine and mirtazapine. Life Sci 2024; 358:123133. [PMID: 39413901 DOI: 10.1016/j.lfs.2024.123133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024]
Abstract
Major Depressive Disorder (MDD) is a very complex disease, challenging to study and manage. The complexities of MDD require extensive research of its mechanisms to develop more effective therapeutic approaches. Crucial in the context of this disease is the role of brain-derived neurotrophic factor (BDNF) signaling pathway. AIM This manuscript aims to explore the complex relationship between MDD and BDNF signaling pathway, focusing on how BDNF is modulated in response to oxidative stress and corticosterone, known to be altered in MDD and contributing to the pathology of the disorder, when treated with scopolamine and mirtazapine. METHODS To assess BDNF levels after the different treatment conditions, rat hippocampal slices and mice primary hippocampus and cortical cell culture were analyzed by immunofluorescence and Western blot. KEY FINDINGS Both mirtazapine and scopolamine under stress conditions induced by hydrogen peroxide (H2O2) and corticosterone, had a significant impact on BDNF levels, and this was distinct in different neuronal models. Mirtazapine, especially when combined with H2O2, altered BDNF expression. Scopolamine when combined with both stressors also altered BDNF levels. However, its effects varied depending on the specific neuronal model and stress condition. In accordance with BDNF results, phosphorylated tropomyosin receptor kinase B (pTrkB) presented increased activation when neuronal cells subjected to stress were treated with mirtazapine or scopolamine. SIGNIFICANCE Collectively, this study highlights the complex connection between these compounds, stress conditions, and BDNF/TrkB modulation, supporting the potential therapeutic effects of scopolamine and mirtazapine in modulating BDNF levels, even in stressful conditions.
Collapse
Affiliation(s)
- Ana Salomé Correia
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Marília Torrado
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Tiago Costa-Coelho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Eva Daniela Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; FEUP-Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
| | - Sara Inteiro-Oliveira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Maria José Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ana Paula Pêgo
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Sofia Duque Santos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal.
| |
Collapse
|
3
|
Daher J. A Potential Link between Myeloperoxidase Modified LDL, Atherosclerosis and Depression. Int J Mol Sci 2024; 25:8805. [PMID: 39201490 PMCID: PMC11354346 DOI: 10.3390/ijms25168805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that involves modified low-density lipoproteins (LDL) which play a pivotal role in the initiation and progression of the disease. Myeloperoxidase oxidized LDL (Mox-LDL) is considered to be the most patho-physiologically relevant type of modified LDL and has been reported to be ubiquitously present in atheroma plaques of patients with atherosclerosis. Besides its involvement in the latter disease state, Mox-LDL has also been shown to be implicated in the pathogenesis of various illnesses including sleep disorders, which are in turn associated with heart disease and depression in many intricate ways. Meanwhile, we have recently shown that lox-1-mediated Mox-LDL signaling modulates neuroserpin activity in endothelial cells, which could have major implications that go beyond the pathophysiology of stroke and cerebrovascular disease (CD). Of note is that tissue plasminogen activator (tPA), which is the main target of neuroserpin in the brain, has a crucial function in the processing of brain-derived neurotrophic factor (BDNF) into its mature form. This factor is known to be involved in major depressive disorder (MDD) development and pathogenesis. Since tPA is more conventionally recognized as being involved in fibrinolytic mechanisms, and its effect on the BDNF system in the context of MDD is still not extensively studied, we speculate that any Mox-LDL-driven change in the activity of tPA in patients with atherosclerosis may lead to a decrease in the production of mature BDNF, resulting in impaired neural plasticity and depression. Deciphering the mechanisms of interaction between those factors could help in better understanding the potentially overlapping pathological mechanisms that regulate disease processes in CD and MDD, supporting the possibility of novel and common therapeutic opportunities for millions of patients worldwide.
Collapse
Affiliation(s)
- Jalil Daher
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El-Koura P.O. Box 100, Lebanon
| |
Collapse
|
4
|
K Soman S, Swain M, Dagda RK. BDNF-TrkB Signaling in Mitochondria: Implications for Neurodegenerative Diseases. Mol Neurobiol 2024:10.1007/s12035-024-04357-4. [PMID: 39030441 DOI: 10.1007/s12035-024-04357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a pivotal role in neuronal development, synaptic plasticity, and overall neuronal health by binding to its receptor, tyrosine receptor kinase B (TrkB). This review delves into the intricate mechanisms through which BDNF-TrkB signaling influences mitochondrial function and potentially influences pathology in neurodegenerative diseases. This review highlights the BDNF-TrkB signaling pathway which regulates mitochondrial bioenergetics, biogenesis, and dynamics, mitochondrial processes vital for synaptic transmission and plasticity. Furthermore, we explore how the BDNF-TrkB-PKA signaling in the cytosol and in mitochondria affects mitochondrial transport and distribution and mitochondrial content, which is crucial for supporting the energy demands of synapses. The dysregulation of this signaling pathway is linked to various neurodegenerative diseases, including Alzheimer's and Parkinson's disease, which are characterized by mitochondrial dysfunction and reduced BDNF expression. By examining seminal studies that have characterized this signaling pathway in health and disease, the present review underscores the potential of enhancing BDNF-TrkB signaling to mitigate mitochondrial dysfunction in neurodegenerative diseases, offering insights into therapeutic strategies to enhance neuronal resilience and function.
Collapse
Affiliation(s)
- Smijin K Soman
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Maryann Swain
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Ruben K Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA.
| |
Collapse
|
5
|
McPherson JI, Prakash Krishnan Muthaiah V, Kaliyappan K, Leddy JJ, Personius KE. Temporal expression of brainstem neurotrophic proteins following mild traumatic brain injury. Brain Res 2024; 1835:148908. [PMID: 38582416 DOI: 10.1016/j.brainres.2024.148908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
BDNF, a neurotrophic factor, and its receptors have been implicated in the pathophysiology of mild traumatic brain injury (mTBI). The brainstem houses many vital functions, that are also associated with signs and symptoms of mTBI, but has been understudied in mTBI animal models. We determined the extent to which neurotrophic protein and associated receptor expression is affected within the brainstem of adult rats following mTBI. Their behavioral function was assessed and temporal expression of the 'negative' regulators of neuronal function (p75, t-TrkB, and pro-BDNF) and 'positive' neuroprotective (FL-TrkB and m-BDNF) protein isoforms were determined via western blot and immunohistochemistry at 1, 3, 7, and 14 post-injury days (PID) following mTBI or sham (control) procedure. Within the brainstem, p75 expression increased at PID 1 vs. sham animals. t-TrkB and pro-BDNF expression increased at PID 7 and 14. The 'positive' protein isoforms of FL-TrkB and m-BDNF expression were increased only at PID 7. The ratio of t-TrkB:FL-TrkB (negative:positive) was substantial across groups and time points, suggesting a negative impact of neurotrophic signaling on neuronal function. Additional NeuN experiments revealed cell death occurring within a subset of neurons within the medulla. While behavioral measures improved by PID 7-14, negative neurotrophic biochemical responses persisted. Despite the assertion that mTBI produces "mild" injury, evidence of cell death was observed in the medulla. Ratios of TrkB and BDNF isoforms with conflicting functions suggest that future work should specifically measure each subtype since they induce opposing downstream effects on neuronal function.
Collapse
Affiliation(s)
- Jacob I McPherson
- Department of Rehabilitation Science, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, United States.
| | - Vijaya Prakash Krishnan Muthaiah
- Department of Rehabilitation Science, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, United States
| | - Kathiravan Kaliyappan
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - John J Leddy
- Department of Orthopaedics and Sports Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - Kirkwood E Personius
- Department of Rehabilitation Science, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
6
|
Aleksandrova E, Dimov D, Tacheva T, Petrova H, Celik K, Vlaykova T. Smoking-Dependent Association of Serum Brain-Derived Neurotrophic Factor with Pulmonary Function Parameters in Chronic Obstructive Pulmonary Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1111. [PMID: 39064540 PMCID: PMC11279016 DOI: 10.3390/medicina60071111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: One of the members of the neurotrophin (NT) family is the brain-derived neurotrophic factor (BDNF). In addition to its role in the nerve system, it has been found to play a role in lung health and diseases. Materials and Methods: The serum concentrations of BDNF were assessed in 57 patients with COPD and in 19 control individuals and the possible associations of BDNF with the spirometric indexes and disease stages were explored. Results: We did not find a significant difference between the serum concentrations of BDNF of patients and controls (p = 0.521). A significant negative correlation of the serum BDNF levels with the age of the patients (Rho = -0.279, p = 0.036) was observed. In addition, a borderline negative correlation with the age of disease onset (Rho= -0.244, p = 0.063) was also found. When analyzing these correlations in different genders, we found stronger statistical significance in male patients (Rho = -0.398, p = 0.009; and Rho = -0.419, p = 0.006), while no such significance was found in females (p = 0.574 and p = 0.342). The analyses of the possible relations of serum BDNF concentration with the spirometric parameters in the whole group of patients did not reveal any significance (p = 0.231 for FEV1%pr. and p = 0.271 for FEV1/FVC%). However, when the patients were dichotomized on the basis of smoking habits, we obtained a strong positive correlation between BDNF and FEV1%pr. (Rho = 0.501, p = 0.048) in non-smokers, but strong negative correlations with FEV1%pr. (Rho = -0.468, p = 0.003) and with FEV1/FVC% (Rho = -0.331, p = 0.040) in ex/current smokers. Non-smokers with moderate disease (GOLD II) had higher BDNF serum concentrations than patients with GOLD stage III/IV (p = 0.031). In ex/current smokers, there was an opposite association (p = 0.045). Conclusions: The results of our study suggest that the expression and secretion of BDNF are changed in COPD, but its effects and functions may differ according to the smoking history of the patients.
Collapse
Affiliation(s)
- Elina Aleksandrova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Dimo Dimov
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Tanya Tacheva
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Hristina Petrova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Kahan Celik
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
| | - Tatyana Vlaykova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria; (D.D.); (T.T.); (H.P.); (K.C.); (T.V.)
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| |
Collapse
|
7
|
Yang F, You H, Mizui T, Ishikawa Y, Takao K, Miyakawa T, Li X, Bai T, Xia K, Zhang L, Pang D, Xu Y, Zhu C, Kojima M, Lu B. Inhibiting proBDNF to mature BDNF conversion leads to ASD-like phenotypes in vivo. Mol Psychiatry 2024:10.1038/s41380-024-02595-5. [PMID: 38762692 DOI: 10.1038/s41380-024-02595-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/20/2024]
Abstract
Autism Spectrum Disorders (ASD) comprise a range of early age-onset neurodevelopment disorders with genetic heterogeneity. Most ASD related genes are involved in synaptic function, which is regulated by mature brain-derived neurotrophic factor (mBDNF) and its precursor proBDNF in a diametrically opposite manner: proBDNF inhibits while mBDNF potentiates synapses. Here we generated a knock-in mouse line (BDNFmet/leu) in which the conversion of proBDNF to mBDNF is attenuated. Biochemical experiments revealed residual mBDNF but excessive proBDNF in the brain. Similar to other ASD mouse models, the BDNFmet/leu mice showed reduced dendritic arborization, altered spines, and impaired synaptic transmission and plasticity in the hippocampus. They also exhibited ASD-like phenotypes, including stereotypical behaviors and deficits in social interaction. Moreover, the plasma proBDNF/mBDNF ratio was significantly increased in ASD patients compared to normal children in a case-control study. Thus, deficits in proBDNF to mBDNF conversion in the brain may contribute to ASD-like behaviors, and plasma proBDNF/mBDNF ratio may be a potential biomarker for ASD.
Collapse
Affiliation(s)
- Feng Yang
- China National Clinical Research Center for Neurological Diseases, Basic and Translational Medicine Center, Beijing Tiantan Hospital, Capital Medical University, 100070, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100070, Beijing, China
| | - He You
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100070, Beijing, China
- School of Pharmaceutical Sciences and IDG/McGovern Institute for Brain Research, Tsinghua University, 100084, Beijing, China
| | - Toshiyuki Mizui
- Core Research for Evolutional Science and Technology (CREST), Kawaguchi, 332-0012, Japan
| | - Yasuyuki Ishikawa
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, 371-0816, Japan
| | - Keizo Takao
- Core Research for Evolutional Science and Technology (CREST), Kawaguchi, 332-0012, Japan
- Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of Toyama, Toyama, 930-0194, Japan
| | - Tsuyoshi Miyakawa
- Core Research for Evolutional Science and Technology (CREST), Kawaguchi, 332-0012, Japan
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Xiaofei Li
- China National Clinical Research Center for Neurological Diseases, Basic and Translational Medicine Center, Beijing Tiantan Hospital, Capital Medical University, 100070, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100070, Beijing, China
| | - Ting Bai
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Kun Xia
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Lingling Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dizhou Pang
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Masami Kojima
- Core Research for Evolutional Science and Technology (CREST), Kawaguchi, 332-0012, Japan.
- Biomedical Department of Applied Bioscience, College of Bioscience and Chemistry, Kanazawa Institute of Technology (KIT), Ishikawa, 924-0838, Japan.
| | - Bai Lu
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100070, Beijing, China.
- School of Pharmaceutical Sciences and IDG/McGovern Institute for Brain Research, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
8
|
Zou Y, Zhang Y, Tu M, Ye Y, Li M, Ran R, Zou Z. Brain-derived neurotrophic factor levels across psychiatric disorders: A systemic review and network meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110954. [PMID: 38286331 DOI: 10.1016/j.pnpbp.2024.110954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/07/2024] [Accepted: 01/24/2024] [Indexed: 01/31/2024]
Abstract
As an important neurotrophic factor in the central nervous system, Brain-derived Neurotrophic Factor (BDNF) has been implicated in the pathophysiology of psychiatric disorders in many studies. However, its value as a biomarker for the diagnosis and differential diagnosis of mental disorders is still controversial, and its change patterns among different mental disorders have not been compared. We conducted a network meta-analysis of BDNF levels in different psychiatric disorders including schizophrenia(SCZ), major depressive disorder(MDD), bipolar disorder(BD), panic disorder(PD), post-traumatic stress disorder(PTSD), obsessive-compulsive disorder(OCD), generalized anxiety disorder(GAD) and insomnia. Studies were identified by searching electronic databases through 31/05/2023. BDNF levels decreased in patients with BD, MDD, OCD, PD, SCZ compared with controls, while significantly increased in patients with PTSD. According to the network meta-analysis, BDNF levels were significantly decreased in MDD and SCZ compared with BD (-2.6, 95% CIs [-5.32 to -0.15] and - 2.68 95% CIs [-5.18 to -0.23] respectively). However, in the traditional meta-analysis, there was a trend towards lower BDNF levels in SCZ compared to BD, with no significant difference (SMD = -0.20, 95% CIs [-0.49 to 0.08]). In conclusion, abnormal BDNF levels have been found in psychiatric disorders, and the changes in peripheral BDNF levels in patients with psychiatric disorders were reconfirmed in this study, which suggests BDNF exhibits promising clinical utility and may hold diagnostic value in distinguishing between MDD and BD.
Collapse
Affiliation(s)
- Yazhu Zou
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Yuan Zhang
- Department of pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Mengtian Tu
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Yu Ye
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Mingmei Li
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Rongting Ran
- Southwest Medical University, Luzhou, Sichuan, China
| | - Zhili Zou
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; Key Laboratory of psychosomatic medicine, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China.
| |
Collapse
|
9
|
Homberg JR, Brivio P, Greven CU, Calabrese F. Individuals being high in their sensitivity to the environment: Are sensitive period changes in play? Neurosci Biobehav Rev 2024; 159:105605. [PMID: 38417743 DOI: 10.1016/j.neubiorev.2024.105605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
All individuals on planet earth are sensitive to the environment, but some more than others. These individual differences in sensitivity to environments are seen across many animal species including humans, and can influence personalities as well as vulnerability and resilience to mental disorders. Yet, little is known about the underlying brain mechanisms. Key genes that contribute to individual differences in environmental sensitivity are the serotonin transporter, dopamine D4 receptor and brain-derived neurotrophic factor genes. By synthesizing neurodevelopmental findings of these genetic factors, and discussing them through the lens of mechanisms related to sensitive periods, which are phases of heightened neuronal plasticity during which a certain network is being finetuned by experiences, we propose that these genetic factors delay but extend postnatal sensitive periods. This may explain why sensitive individuals show behavioral features that are characteristic of a young brain state at the level of sensory information processing, such as reduced filtering or blockade of irrelevant information, resulting in a sensory processing system that 'keeps all options open'.
Collapse
Affiliation(s)
- Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Corina U Greven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry Center, London, United Kingdom
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
10
|
Shimada Y, Kumazoe M, Otsuka Y, Tetsuzen R, Fujimura Y, Tachibana H. Neuroprotective effect of isovaleraldehyde accompanied with upregulation of BDNF and CREB phosphorylation via the PKA pathway. J Nat Med 2024; 78:208-215. [PMID: 38063995 DOI: 10.1007/s11418-023-01763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 11/12/2023] [Indexed: 01/04/2024]
Abstract
Recently, the number of patients diagnosed with dementia has increased. The World Health Organization (WHO) estimates that 50 million patients suffer from dementia. Although several therapeutic strategies have been proposed, currently, there is no curative approach for treating dementia. Neurodegeneration is an irreversible process. As this disease gradually progresses over 15-20 years, a low-cost and sustainable method for preventing these diseases is desired. Cacao nib is consumed in many countries, and a recent clinical study indicated that cocoa intake upregulates brain-derived neurotrophic factor (BDNF), which plays a significant role in memory formation and neuronal cell survival. In the present study, neural cells were treated with cacao nib extract or the 17 characteristic components of cacao nib. Treatment with Cacao nib extract upregulates BDNF mRNA expression. In addition, cacao nib extract elicits the phosphorylation of cAMP-response-element-binding protein (CREB), which regulates the transcription of BDNF. Among the 17 species screened, isovaleraldehyde (IVA), also known as an aroma component of cacao nibs extract, improved BDNF mRNA expression without SH-SY5Y cell toxicity. IVA also promoted CREB phosphorylation through a cAMP-dependent protein kinase (PKA)-dependent mechanism. In conclusion, IVA could be responsible for the BDNF upregulation effect of cacao nib, and IVA upregulated BDNF expression via the PKA-CREB axis.
Collapse
Affiliation(s)
- Yu Shimada
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-Ku, Fukuoka, 819-0395, Japan
| | - Motofumi Kumazoe
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-Ku, Fukuoka, 819-0395, Japan
| | - Yushi Otsuka
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-Ku, Fukuoka, 819-0395, Japan
| | - Rin Tetsuzen
- Meiji Innovation Center, Meiji Co., Ltd, 1-29-1, Nanakuni, Hachioji, Tokyo, 192-0919, Japan
| | - Yoshinori Fujimura
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-Ku, Fukuoka, 819-0395, Japan
| | - Hirofumi Tachibana
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-Ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
11
|
Griego E, Galván EJ. BDNF and Lactate as Modulators of Hippocampal CA3 Network Physiology. Cell Mol Neurobiol 2023; 43:4007-4022. [PMID: 37874456 DOI: 10.1007/s10571-023-01425-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 10/14/2023] [Indexed: 10/25/2023]
Abstract
Growing evidence supports the notion that brain-derived neurotrophic factor (BDNF) and lactate are potent modulators of mammalian brain function. The modulatory actions of those biomolecules influence a wide range of neuronal responses, from the shaping of neuronal excitability to the induction and expression of structural and synaptic plasticity. The biological actions of BDNF and lactate are mediated by their cognate receptors and specific transporters located in the neuronal membrane. Canonical functions of BDNF occur via the tropomyosin-related kinase B receptor (TrkB), whereas lactate acts via monocarboxylate transporters or the hydroxycarboxylic acid receptor 1 (HCAR1). Both receptors are highly expressed in the central nervous system, and some of their physiological actions are particularly well characterized in the hippocampus, a brain structure involved in the neurophysiology of learning and memory. The multifarious neuronal circuitry between the axons of the dentate gyrus granule cells, mossy fibers (MF), and pyramidal neurons of area CA3 is of great interest given its role in specific mnemonic processes and involvement in a growing number of brain disorders. Whereas the modulation exerted by BDNF via TrkB has been extensively studied, the influence of lactate via HCAR1 on the properties of the MF-CA3 circuit is an emerging field. In this review, we discuss the role of both systems in the modulation of brain physiology, with emphasis on the hippocampal CA3 network. We complement this review with original data that suggest cross-modulation is exerted by these two independent neuromodulatory systems.
Collapse
Affiliation(s)
- Ernesto Griego
- Departamento de Farmacobiología, Cinvestav Sur, Mexico City, Mexico.
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, USA.
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, Col. Granjas Coapa, C.P. 14330, Mexico City, Mexico.
| | - Emilio J Galván
- Departamento de Farmacobiología, Cinvestav Sur, Mexico City, Mexico
- Centro de Investigaciones sobre el Envejecimiento, Mexico City, Mexico
| |
Collapse
|
12
|
Cefis M, Chaney R, Wirtz J, Méloux A, Quirié A, Leger C, Prigent-Tessier A, Garnier P. Molecular mechanisms underlying physical exercise-induced brain BDNF overproduction. Front Mol Neurosci 2023; 16:1275924. [PMID: 37868812 PMCID: PMC10585026 DOI: 10.3389/fnmol.2023.1275924] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Accumulating evidence supports that physical exercise (EX) is the most effective non-pharmacological strategy to improve brain health. EX prevents cognitive decline associated with age and decreases the risk of developing neurodegenerative diseases and psychiatric disorders. These positive effects of EX can be attributed to an increase in neurogenesis and neuroplastic processes, leading to learning and memory improvement. At the molecular level, there is a solid consensus to involve the neurotrophin brain-derived neurotrophic factor (BDNF) as the crucial molecule for positive EX effects on the brain. However, even though EX incontestably leads to beneficial processes through BDNF expression, cellular sources and molecular mechanisms underlying EX-induced cerebral BDNF overproduction are still being elucidated. In this context, the present review offers a summary of the different molecular mechanisms involved in brain's response to EX, with a specific focus on BDNF. It aims to provide a cohesive overview of the three main mechanisms leading to EX-induced brain BDNF production: the neuronal-dependent overexpression, the elevation of cerebral blood flow (hemodynamic hypothesis), and the exerkine signaling emanating from peripheral tissues (humoral response). By shedding light on these intricate pathways, this review seeks to contribute to the ongoing elucidation of the relationship between EX and cerebral BDNF expression, offering valuable insights into the potential therapeutic implications for brain health enhancement.
Collapse
Affiliation(s)
- Marina Cefis
- Département des Sciences de l’Activité Physique, Faculté des Sciences, Université du Québec à Montréal, Montreal, QC, Canada
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Remi Chaney
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Julien Wirtz
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Alexandre Méloux
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Clémence Leger
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
- Département Génie Biologique, Institut Universitaire de Technologie, Dijon, France
| |
Collapse
|
13
|
Ziegler T, Tsiountsioura M, Meixner-Goetz L, Cvirn G, Lamprecht M. Polyphenols' Impact on Selected Biomarkers of Brain Aging in Healthy Middle-Aged and Elderly Subjects: A Review of Clinical Trials. Nutrients 2023; 15:3770. [PMID: 37686802 PMCID: PMC10490411 DOI: 10.3390/nu15173770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
With a constantly growing elderly population, incidences of neurodegenerative diseases are also rising and are expected to further increase over the next years, while costing health systems across the world trillions of dollars. Therefore, biomarkers to detect manifestations of brain aging early and interventions to slow down its pace are of great interest. In the last years, the importance of the neurotrophins brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) in the context of cognitive function and the aging brain has increased, besides the already well-established amyloid-beta (Aβ) and tau plaques. Due to their wide range of beneficial health effects as well as their antioxidant and anti-inflammatory properties, a class of secondary plant-metabolites, the so-called polyphenols, gained increasing attention. In this review, we discuss the roles of BDNF, Aβ, NGF, and tau proteins as biomarkers of brain aging and the effect of dietary polyphenol interventions on these biomarkers, assessed via blood analysis, magnetic resonance imaging (MRI), and positron emission tomography (PET).
Collapse
Affiliation(s)
- Tobias Ziegler
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria; (T.Z.); (M.T.); (G.C.)
- Juice Plus+ Science Institute, Memphis, TN 38017, USA;
| | - Melina Tsiountsioura
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria; (T.Z.); (M.T.); (G.C.)
- Juice Plus+ Science Institute, Memphis, TN 38017, USA;
| | | | - Gerhard Cvirn
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria; (T.Z.); (M.T.); (G.C.)
| | - Manfred Lamprecht
- Juice Plus+ Science Institute, Memphis, TN 38017, USA;
- Green Beat Institute of Nutrient Research, 8010 Graz, Austria
| |
Collapse
|
14
|
Anderson C, Hicks AJ, Carmichael J, Burke R, Ponsford J. COMT Val158Met and BDNF Val66Met Single-Nucleotide Polymorphisms Are Not Associated With Emotional Distress One Year After Moderate-Severe Traumatic Brain Injury. Neurotrauma Rep 2023; 4:495-506. [PMID: 37636335 PMCID: PMC10457651 DOI: 10.1089/neur.2023.0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023] Open
Abstract
Emotional distress is a common, but poorly addressed, feature of moderate-severe traumatic brain injury (TBI). Previously identified sociodemographic, psychological, and injury-related factors account for only a small proportion of the variability in emotional distress post-TBI. Genetic factors may help to further understand emotional distress in this population. The catechol-O-methyltransferase (COMT) Val158 and brain-derived neurotrophic factor (BDNF) 66Met single-nucleotide polymorphisms (SNPs) have been identified as possible contributory factors to outcomes after TBI. We investigated whether the COMT Val158 and BDNF 66Met SNPs were associated with emotional distress 1 year after moderate-severe TBI, and whether these associations were moderated by age, sex, and TBI severity (as measured by the duration of post-traumatic amnesia [PTA]). Moderate-severe TBI survivors (COMT, n = 391; BDNF, n = 311) provided saliva samples after admission to a TBI rehabilitation hospital. At a follow-up interview ∼1 year after injury, participants completed a self-report measure of emotional distress (Hospital Anxiety and Depression Scale; HADS). Multiple linear regression models were constructed for each SNP to predict total scores on the HADS. Neither COMT Val158 nor BDNF 66Met carriage status (carrier vs. non-carrier) significantly predicted emotional distress (COMT, p = 0.49; BDNF, p = 0.66). Interactions of SNP × age (COMT, p = 0.90; BDNF, p = 0.93), SNP × sex (COMT, p = 0.09; BDNF, p = 0.60), SNP × injury severity (COMT, p = 0.53; BDNF, p = 0.87), and SNP × sex × age (COMT, p = 0.08; BDNF, p = 0.76) were also non-significant. Our null findings suggest that COMT Val158 and BDNF 66Met SNPs do not aid the prediction of emotional distress 1 year after moderate-severe TBI, neither in isolation nor in interaction with age, sex and injury severity. The reporting of null findings such as ours is important to avoid publication bias and prompt researchers to consider the challenges of single-gene candidate studies in understanding post-TBI outcomes. Analyses in larger samples that incorporate multiple genetic factors and their relevant moderating factors may provide a greater understanding of the role of genetics in post-TBI emotional distress.
Collapse
Affiliation(s)
- Chloe Anderson
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Amelia J. Hicks
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Jai Carmichael
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Richard Burke
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Jennie Ponsford
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
15
|
Nelson ML, Pfeifer JA, Hickey JP, Collins AE, Kalisch BE. Exploring Rosiglitazone's Potential to Treat Alzheimer's Disease through the Modulation of Brain-Derived Neurotrophic Factor. BIOLOGY 2023; 12:1042. [PMID: 37508471 PMCID: PMC10376118 DOI: 10.3390/biology12071042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/24/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that debilitates over 55 million individuals worldwide. Currently, treatments manage and alleviate its symptoms; however, there is still a need to find a therapy that prevents or halts disease progression. Since AD has been labeled as "type 3 diabetes" due to its similarity in pathological hallmarks, molecular pathways, and comorbidity with type 2 diabetes mellitus (T2DM), there is growing interest in using anti-diabetic drugs for its treatment. Rosiglitazone (RSG) is a peroxisome proliferator-activated receptor-gamma agonist that reduces hyperglycemia and hyperinsulinemia and improves insulin signaling. In cellular and rodent models of T2DM-associated cognitive decline and AD, RSG has been reported to improve cognitive impairment and reverse AD-like pathology; however, results from human clinical trials remain consistently unsuccessful. RSG has also been reported to modulate the expression of brain-derived neurotrophic factor (BDNF), a protein that regulates neuroplasticity and energy homeostasis and is implicated in both AD and T2DM. The present review investigates RSG's limitations and potential therapeutic benefits in pre-clinical models of AD through its modulation of BDNF expression.
Collapse
Affiliation(s)
- Mackayla L Nelson
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Julia A Pfeifer
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jordan P Hickey
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Andrila E Collins
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Bettina E Kalisch
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
16
|
Aby K, Antony R, Li Y. ProBDNF Upregulation in Murine Hind Limb Ischemia Reperfusion Injury: A Driver of Inflammation. BIOLOGY 2023; 12:903. [PMID: 37508336 PMCID: PMC10375988 DOI: 10.3390/biology12070903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023]
Abstract
Brain-derived neurotropic factor (BDNF) has been shown to be expressed in many nonneuronal tissues including skeletal muscle. Skeletal muscle BDNF has been studied regarding its function in metabolism and exercise; however, less is known about its role in skeletal muscle injury. The precursor to BDNF, proBDNF, has an unknown role in skeletal muscle. The levels of proBDNF, mature BDNF, and their receptors were compared in the skeletal muscle and brain tissues of C57BL/6J mice. Tourniquet-induced hind limb ischemia-reperfusion injury was used to assess the function of skeletal muscle-derived proBDNF in skeletal muscle injury. Skeletal muscle-specific knockout of BDNF and pharmacological inhibition of p75NTR, the proBDNF receptor, were used to determine the role of proBDNF-p75NTR signaling. We show for the first time that proBDNF is the predominantly expressed form of BDNF in skeletal muscle and that proBDNF is significantly upregulated in skeletal muscle following hind limb ischemia-reperfusion injury. Skeletal muscle-specific knockout of BDNF blunted the inflammatory response in the injured tissue and appears to be mediated by the proBDNF-p75NTR pathway, as shown by the pharmacological inhibition of p75NTR. These findings suggest that skeletal muscle proBDNF plays a critical role in driving the inflammatory response following skeletal muscle injury.
Collapse
Affiliation(s)
| | | | - Yifan Li
- Department of Basic Biomedical Science, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (K.A.); (R.A.)
| |
Collapse
|
17
|
Esvald EE, Tuvikene J, Kiir CS, Avarlaid A, Tamberg L, Sirp A, Shubina A, Cabrera-Cabrera F, Pihlak A, Koppel I, Palm K, Timmusk T. Revisiting the expression of BDNF and its receptors in mammalian development. Front Mol Neurosci 2023; 16:1182499. [PMID: 37426074 PMCID: PMC10325033 DOI: 10.3389/fnmol.2023.1182499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/22/2023] [Indexed: 07/11/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) promotes the survival and functioning of neurons in the central nervous system and contributes to proper functioning of many non-neural tissues. Although the regulation and role of BDNF have been extensively studied, a rigorous analysis of the expression dynamics of BDNF and its receptors TrkB and p75NTR is lacking. Here, we have analyzed more than 3,600 samples from 18 published RNA sequencing datasets, and used over 17,000 samples from GTEx, and ~ 180 samples from BrainSpan database, to describe the expression of BDNF in the developing mammalian neural and non-neural tissues. We show evolutionarily conserved dynamics and expression patterns of BDNF mRNA and non-conserved alternative 5' exon usage. Finally, we also show increasing BDNF protein levels during murine brain development and BDNF protein expression in several non-neural tissues. In parallel, we describe the spatiotemporal expression pattern of BDNF receptors TrkB and p75NTR in both murines and humans. Collectively, our in-depth analysis of the expression of BDNF and its receptors gives insight into the regulation and signaling of BDNF in the whole organism throughout life.
Collapse
Affiliation(s)
- Eli-Eelika Esvald
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
| | - Jürgen Tuvikene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
- dxlabs LLC, Tallinn, Estonia
| | - Carl Sander Kiir
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Annela Avarlaid
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Laura Tamberg
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Alex Sirp
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Anastassia Shubina
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | | | - Indrek Koppel
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | - Tõnis Timmusk
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
| |
Collapse
|
18
|
Çerçi B, Gök A, Akyol A. Brain-derived neurotrophic factor: Its role in energy balance and cancer cachexia. Cytokine Growth Factor Rev 2023; 71-72:105-116. [PMID: 37500391 DOI: 10.1016/j.cytogfr.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 07/29/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in the development of the central and peripheral nervous system during embryogenesis. In the mature central nervous system, BDNF is required for the maintenance and enhancement of synaptic transmissions and the survival of neurons. Particularly, it is involved in the modulation of neurocircuits that control energy balance through food intake, energy expenditure, and locomotion. Regulation of BDNF in the central nervous system is complex and environmental factors affect its expression in murine models which may reflect to phenotype dramatically. Furthermore, BDNF and its high-affinity receptor tropomyosin receptor kinase B (TrkB), as well as pan-neurotrophin receptor (p75NTR) is expressed in peripheral tissues in adulthood and their signaling is associated with regulation of energy balance. BDNF/TrkB signaling is exploited by cancer cells as well and BDNF expression is increased in tumors. Intriguingly, previously demonstrated roles of BDNF in regulation of food intake, adipose tissue and muscle overlap with derangements observed in cancer cachexia. However, data about the involvement of BDNF in cachectic cancer patients and murine models are scarce and inconclusive. In the future, knock-in and/or knock-out experiments with murine cancer models could be helpful to explore potential new roles for BDNF in the development of cancer cachexia.
Collapse
Affiliation(s)
- Barış Çerçi
- Medical School, Hacettepe University, Ankara, Turkey.
| | - Ayşenur Gök
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey; Hacettepe University Transgenic Animal Technologies Research and Application Center, Sıhhiye, Ankara 06100, Turkey
| | - Aytekin Akyol
- Departmant of Pathology, Medical School, Hacettepe University, Ankara, Turkey; Hacettepe University Transgenic Animal Technologies Research and Application Center, Sıhhiye, Ankara 06100, Turkey
| |
Collapse
|
19
|
Nakano S, Kashio S, Nishimura K, Takeishi A, Kosakamoto H, Obata F, Kuranaga E, Chihara T, Yamauchi Y, Isobe T, Miura M. Damage sensing mediated by serine proteases Hayan and Persephone for Toll pathway activation in apoptosis-deficient flies. PLoS Genet 2023; 19:e1010761. [PMID: 37319131 DOI: 10.1371/journal.pgen.1010761] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/25/2023] [Indexed: 06/17/2023] Open
Abstract
The mechanisms by which the innate immune system senses damage have been extensively explored in multicellular organisms. In Drosophila, various types of tissue damage, including epidermal injury, tumor formation, cell competition, and apoptosis deficiency, induce sterile activation of the Toll pathway, a process that requires the use of extracellular serine protease (SP) cascades. Upon infection, the SP Spätzle (Spz)-processing enzyme (SPE) cleaves and activates the Toll ligand Spz downstream of two paralogous SPs, Hayan and Persephone (Psh). However, upon tissue damage, it is not fully understood which SPs establish Spz activation cascades nor what damage-associated molecules can activate SPs. In this study, using newly generated uncleavable spz mutant flies, we revealed that Spz cleavage is required for the sterile activation of the Toll pathway, which is induced by apoptosis-deficient damage of wing epidermal cells in adult Drosophila. Proteomic analysis of hemolymph, followed by experiments with Drosophila Schneider 2 (S2) cells, revealed that among hemolymph SPs, both SPE and Melanization Protease 1 (MP1) have high capacities to cleave Spz. Additionally, in S2 cells, MP1 acts downstream of Hayan and Psh in a similar manner to SPE. Using genetic analysis, we found that the upstream SPs Hayan and Psh contributes to the sterile activation of the Toll pathway. While SPE/MP1 double mutants show more impairment of Toll activation upon infection than SPE single mutants, Toll activation is not eliminated in these apoptosis-deficient flies. This suggests that Hayan and Psh sense necrotic damage, inducing Spz cleavage by SPs other than SPE and MP1. Furthermore, hydrogen peroxide, a representative damage-associated molecule, activates the Psh-Spz cascade in S2 cells overexpressing Psh. Considering that reactive oxygen species (ROS) were detected in apoptosis-deficient wings, our findings highlight the importance of ROS as signaling molecules that induce the activation of SPs such as Psh in response to damage.
Collapse
Affiliation(s)
- Shotaro Nakano
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Soshiro Kashio
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kei Nishimura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Asuka Takeishi
- Neural Circuit of Multisensory Integration RIKEN Hakubi Research Team, RIKEN Center for Brain Science, RIKEN Cluster for Pioneering Research, Wako, Japan
| | - Hina Kosakamoto
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Fumiaki Obata
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Laboratory of Molecular Cell Biology and Development, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Erina Kuranaga
- Laboratory of Histogenetic Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Takahiro Chihara
- Program of Biomedical Science and Program of Basic Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima Japan
| | - Yoshio Yamauchi
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Japan
| | - Toshiaki Isobe
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
20
|
Chiavacci E, Bagnoli S, Cellerino A, Terzibasi Tozzini E. Distribution of Brain-Derived Neurotrophic Factor in the Brain of the Small-Spotted Catshark Scyliorhinus canicula, and Evolution of Neurotrophins in Basal Vertebrates. Int J Mol Sci 2023; 24:ijms24119495. [PMID: 37298444 DOI: 10.3390/ijms24119495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/17/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Neurotrophins (NTFs) are structurally related neurotrophic factors essential for differentiation, survival, neurite outgrowth, and the plasticity of neurons. Abnormalities associated with neurotrophin-signaling (NTF-signaling) were associated with neuropathies, neurodegenerative disorders, and age-associated cognitive decline. Among the neurotrophins, brain-derived neurotrophic factor (BDNF) has the highest expression and is expressed in mammals by specific cells throughout the brain, with particularly high expression in the hippocampus and cerebral cortex. Whole genome sequencing efforts showed that NTF signaling evolved before the evolution of Vertebrates; thus, the shared ancestor of Protostomes, Cyclostomes, and Deuterostomes must have possessed a single ortholog of neurotrophins. After the first round of whole genome duplication that occurred in the last common ancestor of Vertebrates, the presence of two neurotrophins in Agnatha was hypothesized, while the monophyletic group of cartilaginous fishes, or Chondrichthyans, was situated immediately after the second whole genome duplication round that occurred in the last common ancestor of Gnathostomes. Chondrichthyans represent the outgroup of all other living jawed vertebrates (Gnathostomes) and the sister group of Osteichthyans (comprehensive of Actinopterygians and Sarcopterygians). We were able to first identify the second neurotrophin in Agnatha. Secondly, we expanded our analysis to include the Chondrichthyans, with their strategic phylogenetic position as the most basal extant Gnathostome taxon. Results from the phylogenetic analysis confirmed the presence of four neurotrophins in the Chondrichthyans, namely the orthologs of the four mammalian neurotrophins BDNF, NGF, NT-3, and NT-4. We then proceeded to study the expression of BDNF in the adult brain of the Chondrichthyan Scyliorhinus canicula. Our results showed that BDNF is highly expressed in the S. canicula brain and that its expression is highest in the Telencephalon, while the Mesencephalic and Diencephalic areas showed expression of BDNF in isolated and well-defined cell groups. NGF was expressed at much lower levels that could be detected by PCR but not by in situ hybridization. Our results warrant further investigations in Chondrichthyans to characterize the putative ancestral function of neurotrophins in Vertebrates.
Collapse
Affiliation(s)
- Elena Chiavacci
- Biology Laboratory (BIO@SNS), Scuola Normale Superiore, 56126 Pisa, Italy
- Biology and Evolution of Marine Organisms Department (BEOM), Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Sara Bagnoli
- Biology Laboratory (BIO@SNS), Scuola Normale Superiore, 56126 Pisa, Italy
| | - Alessandro Cellerino
- Biology Laboratory (BIO@SNS), Scuola Normale Superiore, 56126 Pisa, Italy
- Fritz Lipmann Institute for Age Research, Leibniz Institute, 07745 Jena, Germany
| | - Eva Terzibasi Tozzini
- Biology and Evolution of Marine Organisms Department (BEOM), Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| |
Collapse
|
21
|
Arévalo JC, Deogracias R. Mechanisms Controlling the Expression and Secretion of BDNF. Biomolecules 2023; 13:biom13050789. [PMID: 37238659 DOI: 10.3390/biom13050789] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/19/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Brain-derived nerve factor (BDNF), through TrkB receptor activation, is an important modulator for many different physiological and pathological functions in the nervous system. Among them, BDNF plays a crucial role in the development and correct maintenance of brain circuits and synaptic plasticity as well as in neurodegenerative diseases. The proper functioning of the central nervous system depends on the available BDNF concentrations, which are tightly regulated at transcriptional and translational levels but also by its regulated secretion. In this review we summarize the new advances regarding the molecular players involved in BDNF release. In addition, we will address how changes of their levels or function in these proteins have a great impact in those functions modulated by BDNF under physiological and pathological conditions.
Collapse
Affiliation(s)
- Juan Carlos Arévalo
- Department of Cell Biology and Pathology, Institute of Neurosciences of Castille and Leon (INCyL), University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Rubén Deogracias
- Department of Cell Biology and Pathology, Institute of Neurosciences of Castille and Leon (INCyL), University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
22
|
Pisani A, Paciello F, Del Vecchio V, Malesci R, De Corso E, Cantone E, Fetoni AR. The Role of BDNF as a Biomarker in Cognitive and Sensory Neurodegeneration. J Pers Med 2023; 13:jpm13040652. [PMID: 37109038 PMCID: PMC10140880 DOI: 10.3390/jpm13040652] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has a crucial function in the central nervous system and in sensory structures including olfactory and auditory systems. Many studies have highlighted the protective effects of BDNF in the brain, showing how it can promote neuronal growth and survival and modulate synaptic plasticity. On the other hand, conflicting data about BDNF expression and functions in the cochlear and in olfactory structures have been reported. Several clinical and experimental research studies showed alterations in BDNF levels in neurodegenerative diseases affecting the central and peripheral nervous system, suggesting that BDNF can be a promising biomarker in most neurodegenerative conditions, including Alzheimer's disease, shearing loss, or olfactory impairment. Here, we summarize current research concerning BDNF functions in brain and in sensory domains (olfaction and hearing), focusing on the effects of the BDNF/TrkB signalling pathway activation in both physiological and pathological conditions. Finally, we review significant studies highlighting the possibility to target BDNF as a biomarker in early diagnosis of sensory and cognitive neurodegeneration, opening new opportunities to develop effective therapeutic strategies aimed to counteract neurodegeneration.
Collapse
Affiliation(s)
- Anna Pisani
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Valeria Del Vecchio
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| | - Rita Malesci
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| | - Eugenio De Corso
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Elena Cantone
- Department of Neuroscience, Reproductive Sciences and Dentistry-ENT Section, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Rita Fetoni
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
23
|
Marques DM, Almeida AS, Oliveira CBA, Machado ACL, Lara MVS, Porciúncula LO. Delayed Outgrowth in Response to the BDNF and Altered Synaptic Proteins in Neurons From SHR Rats. Neurochem Res 2023:10.1007/s11064-023-03917-9. [PMID: 36995561 DOI: 10.1007/s11064-023-03917-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/23/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by inattention, hyperactivity, and impulsivity symptoms. Neuroimaging studies have revealed a delayed cortical and subcortical development pattern in children diagnosed with ADHD. This study followed up on the development in vitro of frontal cortical neurons from Spontaneously hypertensive rats (SHR), an ADHD rat model, and Wistar-Kyoto rats (WKY), control strain, over their time in culture, and in response to BDNF treatment at two different days in vitro (DIV). These neurons were also evaluated for synaptic proteins, brain-derived neurotrophic factor (BDNF), and related protein levels. Frontal cortical neurons from the ADHD rat model exhibited shorter dendrites and less dendritic branching over their time in culture. While pro- and mature BDNF levels were not altered, the cAMP-response element-binding (CREB) decreased at 1 DIV and SNAP-25 decreased at 5 DIV. Different from control cultures, exogenous BDNF promoted less dendritic branching in neurons from the ADHD model. Our data revealed that neurons from the ADHD model showed decreased levels of an important transcription factor at the beginning of their development, and their delayed outgrowth and maturation had consequences in the levels of SNAP-25 and may be associated with less response to BDNF. These findings provide an alternative tool for studies on synaptic dysfunctions in ADHD. They may also offer a valuable tool for investigating drug effects and new treatment opportunities.
Collapse
Affiliation(s)
- Daniela M Marques
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Amanda S Almeida
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Catiane B A Oliveira
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Ana Carolina L Machado
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Marcus Vinícius S Lara
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Lisiane O Porciúncula
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil.
| |
Collapse
|
24
|
Di Donato M, Giovannelli P, Migliaccio A, Castoria G. The nerve growth factor-delivered signals in prostate cancer and its associated microenvironment: when the dialogue replaces the monologue. Cell Biosci 2023; 13:60. [PMID: 36941697 PMCID: PMC10029315 DOI: 10.1186/s13578-023-01008-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/06/2023] [Indexed: 03/22/2023] Open
Abstract
Prostate cancer (PC) represents the most diagnosed and the second most lethal cancer in men worldwide. Its development and progression occur in concert with alterations in the surrounding tumor microenvironment (TME), made up of stromal cells and extracellular matrix (ECM) that dynamically interact with epithelial PC cells affecting their growth and invasiveness. PC cells, in turn, can functionally sculpt the TME through the secretion of various factors, including neurotrophins. Among them, the nerve growth factor (NGF) that is released by both epithelial PC cells and carcinoma-associated fibroblasts (CAFs) triggers the activation of various intracellular signaling cascades, thereby promoting the acquisition of a metastatic phenotype. After many years of investigation, it is indeed well established that aberrations and/or derangement of NGF signaling are involved not only in neurological disorders, but also in the pathogenesis of human proliferative diseases, including PC. Another key feature of cancer progression is the nerve outgrowth in TME and the concept of nerve dependence related to perineural invasion is currently emerging. NGF released by cancer cells can be a driver of tumor neurogenesis and nerves infiltrated in TME release neurotransmitters, which might stimulate the growth and sustainment of tumor cells.In this review, we aim to provide a snapshot of NGF action in the interactions between TME, nerves and PC cells. Understanding the molecular basis of this dialogue might expand the arsenal of therapeutic strategies against this widespread disease.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy.
| | - Pia Giovannelli
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy.
| | - Antimo Migliaccio
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy
| |
Collapse
|
25
|
Kania B, Sotelo A, Ty D, Wisco JJ. The Prevention of Inflammation and the Maintenance of Iron and Hepcidin Homeostasis in the Gut, Liver, and Brain Pathologies. J Alzheimers Dis 2023; 92:769-789. [PMID: 36846996 PMCID: PMC10116142 DOI: 10.3233/jad-220224] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
The human gut microbiome consists of a variety of microorganisms that inhabit the intestinal tract. This flora has recently been shown to play an important role in human disease. The crosstalk between the gut and brain axis has been investigated through hepcidin, derived from both hepatocytes and dendritic cells. Hepcidin could potentially play an anti-inflammatory role in the process of gut dysbiosis through a means of either a localized approach of nutritional immunity, or a systemic approach. Like hepcidin, mBDNF and IL-6 are part of the gut-brain axis: gut microbiota affects their levels of expression, and this relationship is thought to play a role in cognitive function and decline, which could ultimately lead to a number of neurodegenerative diseases such as Alzheimer's disease. This review will focus on the interplay between gut dysbiosis and the crosstalk between the gut, liver, and brain and how this is mediated by hepcidin through different mechanisms including the vagus nerve and several different biomolecules. This overview will also focus on the gut microbiota-induced dysbiotic state on a systemic level, and how gut dysbiosis can contribute to beginnings and the progression of Alzheimer's disease and neuroinflammation.
Collapse
Affiliation(s)
- Barbara Kania
- Department of Anatomy and Neurobiology, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston, MA, USA
| | - Alexis Sotelo
- Department of Anatomy and Neurobiology, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston, MA, USA
| | - Darren Ty
- Department of Anatomy and Neurobiology, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston, MA, USA
| | - Jonathan J Wisco
- Department of Anatomy and Neurobiology, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
26
|
Matsubayashi J, Kawaguchi Y, Kawakami Y, Takei K. Brain-derived neurotrophic factor (BDNF) induces antagonistic action to Nogo signaling by the upregulation of lateral olfactory tract usher substance (LOTUS) expression. J Neurochem 2023; 164:29-43. [PMID: 36448220 DOI: 10.1111/jnc.15732] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Neurons in the central nervous system (CNS) have limited capacity for axonal regeneration after trauma and neurological disorders due to an endogenous nonpermissive environment for axon regrowth in the CNS. Lateral olfactory tract usher substance (LOTUS) contributes to axonal tract formation in the developing brain and axonal regeneration in the adult brain as an endogenous Nogo receptor-1 (NgR1) antagonist. However, how LOTUS expression is regulated remains unclarified. This study examined molecular mechanism of regulation in LOTUS expression and found that brain-derived neurotrophic factor (BDNF) increased LOTUS expression in cultured hippocampal neurons. Exogenous application of BDNF increased LOTUS expression at both mRNA and protein levels in a dose-dependent manner. We also found that pharmacological inhibition with K252a and gene knockdown by siRNA of tropomyosin-related kinase B (TrkB), BDNF receptor suppressed BDNF-induced increase in LOTUS expression. Further pharmacological analysis of the TrkB signaling pathway revealed that BDNF increased LOTUS expression through mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) cascades, but not phospholipase C-γ (PLCγ) cascade. Additionally, treatment with c-AMP response element binding protein (CREB) inhibitor partially suppressed BDNF-induced LOTUS expression. Finally, neurite outgrowth assay in cultured hippocampal neurons revealed that BDNF treatment-induced antagonism for NgR1 by up-regulating LOTUS expression. These findings suggest that BDNF may acts as a positive regulator of LOTUS expression through the TrkB signaling, thereby inducing an antagonistic action for NgR1 function by up-regulating LOTUS expression. Also, BDNF may synergistically affect axon regrowth through the upregulation of LOTUS expression.
Collapse
Affiliation(s)
- Junpei Matsubayashi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| | - Yuki Kawaguchi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| | - Yutaka Kawakami
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan.,Department of Anesthesiology, National Center for Neurology and Psychiatry, Kodaira, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| |
Collapse
|
27
|
Forte ARCC, Lessa PHC, Chaves Filho AJM, Aquino PEAD, Brito LM, Pinheiro LC, Juruena MF, Lucena DFD, de Rezende PHF, de Vasconcelos SMM. Oxidative stress and inflammatory process in borderline personality disorder (BPD): a narrative review. Braz J Med Biol Res 2023; 56:e12484. [PMID: 36946840 PMCID: PMC10021502 DOI: 10.1590/1414-431x2023e12484] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/13/2023] [Indexed: 03/23/2023] Open
Abstract
Borderline personality disorder (BPD) is a severe psychiatric condition that affects up to 2.7% of the population and is highly linked to functional impairment and suicide. Despite its severity, there is a lack of knowledge about its pathophysiology. Studies show genetic influence and childhood violence as factors that may contribute to the development of BPD; however, the involvement of neuroinflammation in BPD remains poorly investigated. This article aimed to explore the pathophysiology of BPD according to the levels of brain-derived neurotrophic factor (BDNF), inflammatory cytokines, and oxidative stress substances that exacerbate neuronal damage. Few articles have been published on this theme. They show that patients with BPD have a lower level of BDNF and a higher level of tumor necrosis factor (TNF)-α and interleukin (IL)-6 in peripheral blood, associated with increased plasma levels of oxidative stress markers, such as malondialdehyde and 8-hydroxy-2-deoxyguanosine. Therefore, more research on the topic is needed, mainly with a pre-clinical and clinical focus.
Collapse
Affiliation(s)
- A R C C Forte
- Laboratório de Neuropsicofarmacologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P H C Lessa
- Curso de Medicina, Departamento de Ciências Biológicas e da Saúde (DCBS), Universidade Federal do Amapá, Macapá, AP, Brasil
| | - A J M Chaves Filho
- Laboratório de Neuropsicofarmacologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P E A de Aquino
- Laboratório de Neuropsicofarmacologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - L M Brito
- Laboratório de Neuropsicofarmacologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - L C Pinheiro
- Laboratório de Neuropsicofarmacologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M F Juruena
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - D F de Lucena
- Laboratório de Neuropsicofarmacologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P H F de Rezende
- Laboratório de Neuropsicofarmacologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - S M M de Vasconcelos
- Laboratório de Neuropsicofarmacologia, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
28
|
de Assis GG, Hoffman JR. The BDNF Val66Met Polymorphism is a Relevant, But not Determinant, Risk Factor in the Etiology of Neuropsychiatric Disorders - Current Advances in Human Studies: A Systematic Review. Brain Plast 2022; 8:133-142. [PMID: 36721394 PMCID: PMC9837733 DOI: 10.3233/bpl-210132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2022] [Indexed: 02/03/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is the brain's most-produced neurotrophin during the lifespan, essentially involved in multiple mechanisms of nervous system development and function. The production/release of BDNF requires multi-stage processing that appears to be regulated at various stages in which the presence of a polymorphism "Val66Met" can exert a critical influence. Aim To synthesize the knowledge on the BDNF Val66Met polymorphism on intracellular processing and function of BDNF. Methods We performed a systematic review and collected all available studies on the post-translation processes of BDNF, regarding the Val66Met polymorphism. Searches were performed up to 21st March 2021. Results Out of 129 eligible papers, 18 studies addressed or had findings relating to BDNF post-translation processes and were included in this review. Discussion Compilation of experimental findings reveals that the Val66Met polymorphism affects BDNF function by slightly altering the processing, distribution, and regulated release of BDNF. Regarding the critical role of pro-BDNF as a pro-apoptotic factor, such alteration might represent a risk for the development of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gilmara Gomes de Assis
- Laboratory of Endocrinology, Brain Institute, Federal University of Rio Grande do Norte, Brazil
- Gdansk University of Physical Education and Sports, Faculty of Physical Education, Gdansk, Poland
| | - Jay R. Hoffman
- Department of Physical Therapy, Ariel University, Ariel, Israel
| |
Collapse
|
29
|
Hammid A, Fallon JK, Lassila T, Vieiro P, Balla A, Gonzalez F, Urtti A, Smith PC, Tolonen A, Honkakoski P. Activity and Expression of Carboxylesterases and Arylacetamide Deacetylase in Human Ocular Tissues. Drug Metab Dispos 2022; 50:1483-1492. [PMID: 36195336 DOI: 10.1124/dmd.122.000993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/23/2022] [Indexed: 11/22/2022] Open
Abstract
As a multitissue organ, the eye possesses unique anatomy and physiology, including differential expression of drug-metabolizing enzymes. Several hydrolytic enzymes that play a major role in drug metabolism and bioactivation of prodrugs have been detected in ocular tissues, but data on their quantitative expression is scarce. Also, many ophthalmic drugs are prone to hydrolysis. Metabolic characterization of individual ocular tissues is useful for the drug development process, and therefore, seven individual ocular tissues from human eyes were analyzed for the activity and expression of carboxylesterases (CESs) and arylacetamide deacetylase (AADAC). Generic and selective human esterase substrates 4-nitrophenyl acetate (most esterases), D-luciferin methyl ester (CES1), fluorescein diacetate and procaine (CES2), and phenacetin (AADAC) were applied to determine the enzymes' specific activities. Enzyme kinetics and inhibition studies were performed with isoform-selective inhibitors digitonin (CES1) and verapamil and diltiazem (CES2). Enzyme contents were determined using quantitative targeted proteomics, and CES2 expression was confirmed by western blotting. The expression and activity of human CES1 among ocular tissues varied by >10-fold, with the highest levels found in the retina and iris-ciliary body. In contrast, human CES2 expression appeared lower and more similar between tissues, whereas AADAC could not be detected. Inhibition studies showed that hydrolysis of fluorescein diacetate is also catalyzed by enzymes other than CES2. This study provides, for the first time, quantitative information on the tissue-dependent expression of human ocular esterases, which can be useful for the development of ocular drugs, prodrugs, and in pharmacokinetic modeling of the eye. SIGNIFICANCE STATEMENT: Novel and comprehensive data on the protein expression and activities of carboxylesterases from individual human eye tissues are generated. In combination with previous reports on preclinical species, this study will improve the understanding of interspecies differences in ocular drug metabolism and aid the development of ocular pharmacokinetics models.
Collapse
Affiliation(s)
- Anam Hammid
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - John K Fallon
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Toni Lassila
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Paula Vieiro
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Anusha Balla
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Francisco Gonzalez
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Arto Urtti
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Philip C Smith
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Ari Tolonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland (A.H., A.B., A.U., P.H.); Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (J.K.F., P.C.S.); Admescope Ltd, Oulu, Finland (T.L., A.T.); Biobank at the University Hospital at Santiago de Compostela, Santiago de Compostela, Spain (P.V.); Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain (F.G.); Service of Ophthalmology, University Hospital of Santiago de Compostela and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela, Santiago de Compostela (F.G.); and Faculty of Pharmacy, University of Helsinki, Helsinki, Finland (A.U.)
| |
Collapse
|
30
|
Li Y, Li F, Qin D, Chen H, Wang J, Wang J, Song S, Wang C, Wang Y, Liu S, Gao D, Wang ZH. The role of brain derived neurotrophic factor in central nervous system. Front Aging Neurosci 2022; 14:986443. [PMID: 36158555 PMCID: PMC9493475 DOI: 10.3389/fnagi.2022.986443] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/23/2022] [Indexed: 11/15/2022] Open
Abstract
Brain derived neurotrophic factor (BDNF) has multiple biological functions which are mediated by the activation of two receptors, tropomyosin receptor kinase B (TrkB) receptor and the p75 neurotrophin receptor, involving in physiological and pathological processes throughout life. The diverse presence and activity of BDNF indicate its potential role in the pathogenesis, progression and treatment of both neurological and psychiatric disorders. This review is to provide a comprehensive assessment of the current knowledge and future directions in BDNF-associated research in the central nervous system (CNS), with an emphasis on the physiological and pathological functions of BDNF as well as its potential treatment effects in CNS diseases, including depression, Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, multiple sclerosis, and cerebral ischemic stroke.
Collapse
Affiliation(s)
- Yiyi Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dongdong Qin
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyu Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jianhao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiabei Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shafei Song
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yamei Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Songyan Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dandan Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhi-Hao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Zhi-Hao Wang,
| |
Collapse
|
31
|
Zhang Y, Gao X, Bai X, Yao S, Chang YZ, Gao G. The emerging role of furin in neurodegenerative and neuropsychiatric diseases. Transl Neurodegener 2022; 11:39. [PMID: 35996194 PMCID: PMC9395820 DOI: 10.1186/s40035-022-00313-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Furin is an important mammalian proprotein convertase that catalyzes the proteolytic maturation of a variety of prohormones and proproteins in the secretory pathway. In the brain, the substrates of furin include the proproteins of growth factors, receptors and enzymes. Emerging evidence, such as reduced FURIN mRNA expression in the brains of Alzheimer's disease patients or schizophrenia patients, has implicated a crucial role of furin in the pathophysiology of neurodegenerative and neuropsychiatric diseases. Currently, compared to cancer and infectious diseases, the aberrant expression of furin and its pharmaceutical potentials in neurological diseases remain poorly understood. In this article, we provide an overview on the physiological roles of furin and its substrates in the brain, summarize the deregulation of furin expression and its effects in neurodegenerative and neuropsychiatric disorders, and discuss the implications and current approaches that target furin for therapeutic interventions. This review may expedite future studies to clarify the molecular mechanisms of furin deregulation and involvement in the pathogenesis of neurodegenerative and neuropsychiatric diseases, and to develop new diagnosis and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xiaoqin Gao
- Shijiazhuang People's Hospital, Hebei Medical University, Shijiazhuang, 050027, China
| | - Xue Bai
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Shanshan Yao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
32
|
Zhang B, Guo X. Electroacupuncture promotes nerve regeneration and functional recovery in rats with spinal cord contusion through the coordinate interaction of CD4 and BDNF. IBRAIN 2022; 8:285-301. [PMID: 37786738 PMCID: PMC10529162 DOI: 10.1002/ibra.12055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 10/04/2023]
Abstract
To explore the effect of electroacupuncture on spinal cord injury (SCI) involving immune-related factors and regeneration-related factors in rats. The model of spinal cord contusion was established by PCI 3000 instrument. Two types of acupuncture points were selected for electroacupuncture treatment on rats. The rats were tested once a week, and the fiber remodeling was detected by magnetic resonance imaging. Transcriptome sequencing was performed on spinal scar samples. Using Python to write code, statistical analysis and bioinformatics analysis of the correlation between transcriptome sequencing data and fiber reconstruction results are carried out. Lastly, the expression of CD4 and brain-derived neurotrophic factor (BDNF) in spinal cord scar was verified by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). Electroacupuncture exhibited a positive effect on the recovery of motor function in rats after SCI. Bioinformatics analysis found a direct interaction between CD4 and BDNF. Transcriptome sequencing and PCR results verified that electroacupuncture significantly reduced the expression of CD4, and increased significantly the expression of BDNF, simultaneously corresponding to nerve regeneration in rats with SCI. Our results showed that electroacupuncture intervention in SCI rats improves neural behavior via inhibiting the expression of CD4 and increasing the expression of BDNF.
Collapse
Affiliation(s)
- Bao‐Lei Zhang
- Department of AnatomyJinzhou Medical UniversityJinzhouLiaoningChina
- Department of Experimental ZoologyKunming Medical UniversityKunmingYunnanChina
| | - Xi‐Liang Guo
- Department of Experimental ZoologyKunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
33
|
Rentería I, García-Suárez PC, Fry AC, Moncada-Jiménez J, Machado-Parra JP, Antunes BM, Jiménez-Maldonado A. The Molecular Effects of BDNF Synthesis on Skeletal Muscle: A Mini-Review. Front Physiol 2022; 13:934714. [PMID: 35874524 PMCID: PMC9306488 DOI: 10.3389/fphys.2022.934714] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
The brain-derived neurotrophic factor (BDNF) is a member of the nerve growth factor family which is generated mainly by the brain. Its main role involve synaptic modulation, neurogenesis, neuron survival, immune regulation, myocardial contraction, and angiogenesis in the brain. Together with the encephalon, some peripheral tissues synthesize BDNF like skeletal muscle. On this tissue, this neurotrophin participates on cellular mechanisms related to muscle function maintenance and plasticity as reported on recent scientific works. Moreover, during exercise stimuli the BDNF contributes directly to strengthening neuromuscular junctions, muscle regeneration, insulin-regulated glucose uptake and β-oxidation processes in muscle tissue. Given its vital relevance on many physiological mechanisms, the current mini-review focuses on discussing up-to-date knowledge about BDNF production in skeletal muscle and how this neurotrophin impacts skeletal muscle biology.
Collapse
Affiliation(s)
- I Rentería
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - P C García-Suárez
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico.,Department of Health, Sports and Exercise Sciences, University of Kansas, Lawrence, KS, United States
| | - A C Fry
- Department of Health, Sports and Exercise Sciences, University of Kansas, Lawrence, KS, United States
| | - J Moncada-Jiménez
- Human Movement Sciences Research Center (CIMOHU), University of Costa Rica, San José, Costa Rica
| | - J P Machado-Parra
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - B M Antunes
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - A Jiménez-Maldonado
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| |
Collapse
|
34
|
Schirò G, Iacono S, Ragonese P, Aridon P, Salemi G, Balistreri CR. A Brief Overview on BDNF-Trk Pathway in the Nervous System: A Potential Biomarker or Possible Target in Treatment of Multiple Sclerosis? Front Neurol 2022; 13:917527. [PMID: 35911894 PMCID: PMC9332890 DOI: 10.3389/fneur.2022.917527] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/01/2022] [Indexed: 01/09/2023] Open
Abstract
The growing incidence of neurodegenerative disorders in our populations is leading the research to identify potential biomarkers and targets for facilitating their early management and treatments. Biomarkers represent the crucial indicators of both physiological and pathological processes. Specific changes in molecular and cellular mechanisms of physiological processes result in biochemical alterations at systemic level, which can give us comprehensive information regarding the nature of any disease. In addition, any disease biomarker should be specific and reliable, able to consent of distinguishing the physiological condition of a tissue, organ, or system from disease, and be diverse among the various diseases, or subgroups or phenotypes of them. Accordingly, biomarkers can predict chances for diseases, facilitate their early diagnosis, and set guidelines for the development of new therapies for treating diseases and disease-making process. Here, we focus our attention on brain neurotrophic factor (BDNF)–tropomyosin receptor kinase (Trk) pathway, describing its multiple roles in the maintenance of central nervous system (CNS) health, as well as its implication in the pathogenesis of multiple sclerosis (MS). In addition, we also evidence the features of such pathway, which make of it a potential MS biomarker and therapeutic target.
Collapse
Affiliation(s)
- Giuseppe Schirò
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Salvatore Iacono
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Paolo Ragonese
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Paolo Ragonese
| | - Paolo Aridon
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giuseppe Salemi
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Giuseppe Salemi
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
- *Correspondence: Carmela Rita Balistreri ; orcid.org/0000-0002-5393-1007
| |
Collapse
|
35
|
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
|
36
|
Recent Advances in the Molecular and Cellular Mechanisms of gp120-Mediated Neurotoxicity. Cells 2022; 11:cells11101599. [PMID: 35626635 PMCID: PMC9139548 DOI: 10.3390/cells11101599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 02/01/2023] Open
Abstract
Axonal degeneration and loss of synapses are often seen in different brain areas of people living with human immunodeficiency virus (HIV). Nevertheless, the underlying causes of the pathological alterations observed in these individuals are poorly comprehended, considering that HIV does not infect neurons. Experimental data have shown that viral proteins, including the envelope protein gp120, cause synaptic pathology followed by neuronal cell death. These neurotoxic effects on synapses could be the result of a variety of mechanisms that decrease synaptic plasticity. In this paper, we will briefly present new emerging concepts connected with the ability of gp120 to promote the degeneration of synapses by either directly damaging the axonal cytoskeleton and/or the indirect activation of the p75 neurotrophin receptor death domain in dendrites.
Collapse
|
37
|
Jin T, Zhang Y, Botchway BOA, Zhang J, Fan R, Zhang Y, Liu X. Curcumin can improve Parkinson's disease via activating BDNF/PI3k/Akt signaling pathways. Food Chem Toxicol 2022; 164:113091. [PMID: 35526734 DOI: 10.1016/j.fct.2022.113091] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023]
Abstract
Parkinson's disease is a common progressive neurodegenerative disease, and presently has no curative agent. Curcumin, as one of the natural polyphenols, has great potential in neurodegenerative diseases and other different pathological settings. The brain-derived neurotrophic factor (BDNF) and phosphatidylinositol 3 kinase (PI3k)/protein kinase B (Akt) signaling pathways are significantly involved nerve regeneration and anti-apoptotic activities. Currently, relevant studies have confirmed that curcumin has an optimistic impact on neuroprotection via regulating BDNF and PI3k/Akt signaling pathways in neurodegenerative disease. Here, we summarized the relationship between BDNF and PI3k/Akt signaling pathway, the main biological functions and neuroprotective effects of curcumin via activating BDNF and PI3k/Akt signaling pathways in Parkinson's disease. This paper illustrates that curcumin, as a neuroprotective agent, can delay the progression of Parkinson's disease by protecting nerve cells.
Collapse
Affiliation(s)
- Tian Jin
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Zhang
- Department of Pharmacology, Medical College, Shaoxing University, Zhejiang, China
| | - Ruihua Fan
- School of Life Science, Shaoxing University, Zhejiang, China
| | - Yufeng Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China.
| |
Collapse
|
38
|
Tocotrienols Attenuate White Adipose Tissue Accumulation and Improve Serum Cholesterol Concentration in High-Fat Diet-Treated Mice. Molecules 2022; 27:molecules27072188. [PMID: 35408585 PMCID: PMC9000465 DOI: 10.3390/molecules27072188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 12/04/2022] Open
Abstract
Tocotrienols (T3s), which are vitamin E homologs, have not only antioxidant function but also inhibitory effects on body weight gain and hepatic lipid droplet accumulation. However, the mechanisms of the anti-obesity effects of T3s are not yet understood. In this study, C57BL/6 mice were fed a high-fat diet in the presence or absence of T3s. Treatment with T3s inhibited white adipose tissue accumulation and elevation of serum cholesterol concentrations. Additionally, to clarify the relationship between obesity-induced cognitive dysfunction and the neuroprotective effect of T3s, cognitive function, brain oxidation, and protein expression levels of brain-derived neurotrophic factor (BDNF), which is strongly involved in neuronal growth and differentiation, were measured. Although mice behaviors were improved by oral T3 intake, there were no significant differences in brain oxidation levels and BDNF expression. These results suggest that T3s attenuate obesity via inhibition of body fat and serum cholesterol increase.
Collapse
|
39
|
Canton-Martínez E, Rentería I, García-Suárez PC, Moncada-Jiménez J, Machado-Parra JP, Lira FS, Johnson DK, Jiménez-Maldonado A. Concurrent Training Increases Serum Brain-Derived Neurotrophic Factor in Older Adults Regardless of the Exercise Frequency. Front Aging Neurosci 2022; 14:791698. [PMID: 35330706 PMCID: PMC8940272 DOI: 10.3389/fnagi.2022.791698] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/09/2022] [Indexed: 12/16/2022] Open
Abstract
Background Human brain function declines with aging. In this sense, exercise-based interventions has a promising effect on brain plasticity for older adults. Serum brain-derived neurotrophic factor (BDNF) is a positive biomarker for brain neuroplasticity in healthy older adults also modified by exercise training. Selected features of the exercise prescription for improving brain health are missing; therefore, the aim of this study was to determine the effects of concurrent exercise training frequency on serum BDNF levels in healthy older adults. Methods Nineteen volunteers (age: 65 ± 4 year; body mass index: 28.0 ± 4.5 kg/m2) completed either a three times/week (3-t/w) (n = 8) or five times/week (5-t/w) (n = 11) concurrent exercise program. The exercise program lasted 11 weeks and all exercise sessions were performed for 50 min at moderate intensity. Serum BDNF, body composition, cardiovascular, and physical fitness variables were assessed before and after the exercise training program. Results Regardless of the group, the serum BDNF increased following the intervention (p < 0.001), and there were no significant group (p = 0.827) or interaction (p = 0.063) effects. The maximal oxygen consumption (VO2max) increased regardless of the group (p = 0.007), with a non-significant group (p = 0.722) or interaction (p = 0.223) effects. Upper- and lower-body strength increased in both groups (p = 0.003); however, there was no effect of the training frequency (p = 0.53). For the skeletal muscle mass, there was a trend in the interaction effect (p = 0.053). Finally, the body fat percentage was unchanged. Conclusion Eleven weeks of combined exercise training increased serum BDNF levels in healthy older adults, a response independent of the training frequency. The overall fitness level improved similarly in both exercise groups. These data reveal that a minimal dosage of concurrent exercise enhance functional capacity and a brain health biomarker in older adults.
Collapse
Affiliation(s)
| | - Iván Rentería
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - Patricia C. García-Suárez
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
- Department of Health, Sports and Exercise Sciences, University of Kansas, Lawrence, KS, United States
| | - José Moncada-Jiménez
- Human Movement Sciences Research Center (CIMOHU), University of Costa Rica, San Jose, Costa Rica
| | | | - Fabio Santos Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, Paulista State University, UNESP, Presidente Prudente, Brazil
| | - David K. Johnson
- Department of Neurology, University of California, Davis, Davis, CA, United States
| | - Alberto Jiménez-Maldonado
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
- *Correspondence: Alberto Jiménez-Maldonado,
| |
Collapse
|
40
|
Fading memories in aging and neurodegeneration: Is p75 neurotrophin receptor a culprit? Ageing Res Rev 2022; 75:101567. [PMID: 35051645 DOI: 10.1016/j.arr.2022.101567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/12/2021] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
Aging and age-related neurodegenerative diseases have become one of the major concerns in modern times as cognitive abilities tend to decline when we get older. It is well known that the main cause of this age-related cognitive deficit is due to aberrant changes in cellular, molecular circuitry and signaling pathways underlying synaptic plasticity and neuronal connections. The p75 neurotrophin receptor (p75NTR) is one of the important mediators regulating the fate of the neurons in the nervous system. Its importance in neuronal apoptosis is well documented. However, the mechanisms involving the regulation of p75NTR in synaptic plasticity and cognitive function remain obscure, although cognitive impairment has been associated with a higher expression of p75NTR in neurons. In this review, we discuss the current understanding of how neurons are influenced by p75NTR function to maintain normal neuronal synaptic strength and connectivity, particularly to support learning and memory in the hippocampus. We then discuss the age-associated alterations in neurophysiological mechanisms of synaptic plasticity and cognitive function. Furthermore, we also describe current evidence that has begun to elucidate how p75NTR regulates synaptic changes in aging and age-related neurodegenerative diseases, focusing on the hippocampus. Elucidating the role that p75NTR signaling plays in regulating synaptic plasticity will contribute to a better understanding of cognitive processes and pathological conditions. This will in turn provide novel approaches to improve therapies for the treatment of neurological diseases in which p75NTR dysfunction has been demonstrated.
Collapse
|
41
|
Consequences of the rs6265 (Val66Met) polymorphism in the BDNF gene in selected mental disorders and sport. CURRENT PROBLEMS OF PSYCHIATRY 2022. [DOI: 10.2478/cpp-2022-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Introduction: Brain-derived neurotrophic factor (BDNF) is a polypeptide of 247 amino acid residues and is widely distributed throughout the central nervous system of the CNS. It plays an important role in the survival, differentiation, growth, and development of neurons in the central nervous system. The human BDNF gene is located on chromosome 11 in the p13-14 region and covers approximately 70 kb. The gene has a complex structure as it consists of 11 exons (I-IX, plus Vh and VIIIh) and nine functional promoters. BDNF expression in the brain is relatively low but it is found in most major regions of the brain.
Material and methods: The gene encoding the brain-derived neurotrophic factor BDNF has many polymorphisms, but one of them mainly attracts the attention of researchers. This is a common, non-conservative polymorphism - rs6265 - a single nucleotide SNP polymorphism that results in an amino acid change – valine (Val) to methionine (Met) - at codon 66.
Results: Polymorphism rs6265 is associated with many neuropsychiatric disorders, including depression or a higher risk of addiction, but it also determines other features, such as e.g. sports performance. Few studies are investigating the relationship between rs6265 polymorphism and predisposition to play sports.
Conclusions: The results on the effect of rs6265 BDNF polymorphic variants on the risk of depression and addition are inconsistent, indicating a significant association in some studies and none in others. Therefore, more studies are needed to determine how rs6265 affects gene expression and function.
Collapse
|
42
|
Moretti J, Rodger J. A little goes a long way: Neurobiological effects of low intensity rTMS and implications for mechanisms of rTMS. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100033. [PMID: 36685761 PMCID: PMC9846462 DOI: 10.1016/j.crneur.2022.100033] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/26/2022] [Accepted: 02/15/2022] [Indexed: 01/25/2023] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a widespread technique in neuroscience and medicine, however its mechanisms are not well known. In this review, we consider intensity as a key therapeutic parameter of rTMS, and review the studies that have examined the biological effects of rTMS using magnetic fields that are orders of magnitude lower that those currently used in the clinic. We discuss how extensive characterisation of "low intensity" rTMS has set the stage for translation of new rTMS parameters from a mechanistic evidence base, with potential for innovative and effective therapeutic applications. Low-intensity rTMS demonstrates neurobiological effects across healthy and disease models, which include depression, injury and regeneration, abnormal circuit organisation, tinnitus etc. Various short and long-term changes to metabolism, neurotransmitter release, functional connectivity, genetic changes, cell survival and behaviour have been investigated and we summarise these key changes and the possible mechanisms behind them. Mechanisms at genetic, molecular, cellular and system levels have been identified with evidence that low-intensity rTMS and potentially rTMS in general acts through several key pathways to induce changes in the brain with modulation of internal calcium signalling identified as a major mechanism. We discuss the role that preclinical models can play to inform current clinical research as well as uncover new pathways for investigation.
Collapse
Affiliation(s)
- Jessica Moretti
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia,Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia,Perron Institute for Neurological and Translational Science, Perth, WA, Australia,Corresponding author. School of Biological Sciences M317, The University of Western Australia, 35 Stirling Highway, Crawley WA, 6009, Australia.
| |
Collapse
|
43
|
Gao L, Zhang Y, Sterling K, Song W. Brain-derived neurotrophic factor in Alzheimer's disease and its pharmaceutical potential. Transl Neurodegener 2022; 11:4. [PMID: 35090576 PMCID: PMC8796548 DOI: 10.1186/s40035-022-00279-0] [Citation(s) in RCA: 164] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/01/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic abnormalities are a cardinal feature of Alzheimer's disease (AD) that are known to arise as the disease progresses. A growing body of evidence suggests that pathological alterations to neuronal circuits and synapses may provide a mechanistic link between amyloid β (Aβ) and tau pathology and thus may serve as an obligatory relay of the cognitive impairment in AD. Brain-derived neurotrophic factors (BDNFs) play an important role in maintaining synaptic plasticity in learning and memory. Considering AD as a synaptic disorder, BDNF has attracted increasing attention as a potential diagnostic biomarker and a therapeutical molecule for AD. Although depletion of BDNF has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation and neuronal apoptosis, the exact mechanisms underlying the effect of impaired BDNF signaling on AD are still unknown. Here, we present an overview of how BDNF genomic structure is connected to factors that regulate BDNF signaling. We then discuss the role of BDNF in AD and the potential of BDNF-targeting therapeutics for AD.
Collapse
Affiliation(s)
- Lina Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
44
|
Eggert S, Kins S, Endres K, Brigadski T. Brothers in arms: proBDNF/BDNF and sAPPα/Aβ-signaling and their common interplay with ADAM10, TrkB, p75NTR, sortilin, and sorLA in the progression of Alzheimer's disease. Biol Chem 2022; 403:43-71. [PMID: 34619027 DOI: 10.1515/hsz-2021-0330] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is an important modulator for a variety of functions in the central nervous system (CNS). A wealth of evidence, such as reduced mRNA and protein level in the brain, cerebrospinal fluid (CSF), and blood samples of Alzheimer's disease (AD) patients implicates a crucial role of BDNF in the progression of this disease. Especially, processing and subcellular localization of BDNF and its receptors TrkB and p75 are critical determinants for survival and death in neuronal cells. Similarly, the amyloid precursor protein (APP), a key player in Alzheimer's disease, and its cleavage fragments sAPPα and Aβ are known for their respective roles in neuroprotection and neuronal death. Common features of APP- and BDNF-signaling indicate a causal relationship in their mode of action. However, the interconnections of APP- and BDNF-signaling are not well understood. Therefore, we here discuss dimerization properties, localization, processing by α- and γ-secretase, relevance of the common interaction partners TrkB, p75, sorLA, and sortilin as well as shared signaling pathways of BDNF and sAPPα.
Collapse
Affiliation(s)
- Simone Eggert
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Tanja Brigadski
- Department of Informatics and Microsystem Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| |
Collapse
|
45
|
Porter GA, O’Connor JC. Brain-derived neurotrophic factor and inflammation in depression: Pathogenic partners in crime? World J Psychiatry 2022; 12:77-97. [PMID: 35111580 PMCID: PMC8783167 DOI: 10.5498/wjp.v12.i1.77] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/21/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Major depressive disorder is a debilitating disorder affecting millions of people each year. Brain-derived neurotrophic factor (BDNF) and inflammation are two prominent biologic risk factors in the pathogenesis of depression that have received considerable attention. Many clinical and animal studies have highlighted associations between low levels of BDNF or high levels of inflammatory markers and the development of behavioral symptoms of depression. However, less is known about potential interaction between BDNF and inflammation, particularly within the central nervous system. Emerging evidence suggests that there is bidirectional regulation between these factors with important implications for the development of depressive symptoms and anti-depressant response. Elevated levels of inflammatory mediators have been shown to reduce expression of BDNF, and BDNF may play an important negative regulatory role on inflammation within the brain. Understanding this interaction more fully within the context of neuropsychiatric disease is important for both developing a fuller understanding of biological pathogenesis of depression and for identifying novel therapeutic opportunities. Here we review these two prominent risk factors for depression with a particular focus on pathogenic implications of their interaction.
Collapse
Affiliation(s)
- Grace A Porter
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Jason C O’Connor
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX 78229, United States
- Audie L. Murphy VA Hospital, South Texas Veterans Health System, San Antonio, TX 78229, United States
| |
Collapse
|
46
|
Cefis M, Chaney R, Quirié A, Santini C, Marie C, Garnier P, Prigent-Tessier A. Endothelial cells are an important source of BDNF in rat skeletal muscle. Sci Rep 2022; 12:311. [PMID: 35013359 PMCID: PMC8748777 DOI: 10.1038/s41598-021-03740-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/09/2021] [Indexed: 12/27/2022] Open
Abstract
BDNF (brain-derived neurotrophic factor) is present in skeletal muscle, controlling muscular metabolism, strength and regeneration processes. However, there is no consensus on BDNF cellular source. Furthermore, while endothelial tissue expresses BDNF in large amount, whether endothelial cells inside muscle expressed BDNF has never been explored. The aim of the present study was to provide a comprehensive analysis of BDNF localization in rat skeletal muscle. Cellular localization of BDNF and activated Tropomyosin-related kinase B (TrkB) receptors was studied by immunohistochemical analysis on soleus (SOL) and gastrocnemius (GAS). BDNF and activated TrkB levels were also measured in muscle homogenates using Western blot analysis and/or Elisa tests. The results revealed BDNF immunostaining in all cell types examined with a prominent staining in endothelial cells and a stronger staining in type II than type I muscular fibers. Endothelial cells but not other cells displayed easily detectable activated TrkB receptor expression. Levels of BDNF and activated TrkB receptors were higher in SOL than GAS. In conclusion, endothelial cells are an important and still unexplored source of BDNF present in skeletal muscle. Endothelial BDNF expression likely explains why oxidative muscle exhibits higher BDNF levels than glycolytic muscle despite higher the BDNF expression by type II fibers.
Collapse
Affiliation(s)
- Marina Cefis
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Remi Chaney
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Clélia Santini
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Christine Marie
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
- Département Génie Biologique, IUT, 21000, Dijon, France
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France.
- UFR Des Sciences de Santé, 7 boulevard Jeanne d'Arc, 21078, Dijon, France.
| |
Collapse
|
47
|
Cade S, Zhou XF, Bobrovskaya L. The role of brain-derived neurotrophic factor and the neurotrophin receptor p75NTR in age-related brain atrophy and the transition to Alzheimer's disease. Rev Neurosci 2022; 33:515-529. [PMID: 34982865 DOI: 10.1515/revneuro-2021-0111] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/11/2021] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease is a neurodegenerative condition that is potentially mediated by synaptic dysfunction before the onset of cognitive impairments. The disease mostly affects elderly people and there is currently no therapeutic which halts its progression. One therapeutic strategy for Alzheimer's disease is to regenerate lost synapses by targeting mechanisms involved in synaptic plasticity. This strategy has led to promising drug candidates in clinical trials, but further progress needs to be made. An unresolved problem of Alzheimer's disease is to identify the molecular mechanisms that render the aged brain susceptible to synaptic dysfunction. Understanding this susceptibility may identify drug targets which could halt, or even reverse, the disease's progression. Brain derived neurotrophic factor is a neurotrophin expressed in the brain previously implicated in Alzheimer's disease due to its involvement in synaptic plasticity. Low levels of the protein increase susceptibility to the disease and post-mortem studies consistently show reductions in its expression. A desirable therapeutic approach for Alzheimer's disease is to stimulate the expression of brain derived neurotrophic factor and potentially regenerate lost synapses. However, synthesis and secretion of the protein are regulated by complex activity-dependent mechanisms within neurons, which makes this approach challenging. Moreover, the protein is synthesised as a precursor which exerts the opposite effect of its mature form through the neurotrophin receptor p75NTR. This review will evaluate current evidence on how age-related alterations in the synthesis, processing and signalling of brain derived neurotrophic factor may increase the risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Shaun Cade
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| |
Collapse
|
48
|
Pleiotropic effects of BDNF on the cerebellum and hippocampus: Implications for neurodevelopmental disorders. Neurobiol Dis 2022; 163:105606. [PMID: 34974125 DOI: 10.1016/j.nbd.2021.105606] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 12/17/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the most studied neurotrophins in the mammalian brain, essential not only to the development of the central nervous system but also to synaptic plasticity. BDNF is present in various brain areas, but highest levels of expression are seen in the cerebellum and hippocampus. After birth, BDNF acts in the cerebellum as a mitogenic and chemotactic factor, stimulating the cerebellar granule cell precursors to proliferate, migrate and maturate, while in the hippocampus BDNF plays a fundamental role in synaptic transmission and plasticity, representing a key regulator for the long-term potentiation, learning and memory. Furthermore, the expression of BDNF is highly regulated and changes of its expression are associated with both physiological and pathological conditions. The purpose of this review is to provide an overview of the current state of knowledge on the BDNF biology and its neurotrophic role in the proper development and functioning of neurons and synapses in two important brain areas of postnatal neurogenesis, the cerebellum and hippocampus. Dysregulation of BDNF expression and signaling, resulting in alterations in neuronal maturation and plasticity in both systems, is a common hallmark of several neurodevelopmental diseases, such as autism spectrum disorder, suggesting that neuronal malfunction present in these disorders is the result of excessive or reduced of BDNF support. We believe that the more the relevance of the pathophysiological actions of BDNF, and its downstream signals, in early postnatal development will be highlighted, the more likely it is that new neuroprotective therapeutic strategies will be identified in the treatment of various neurodevelopmental disorders.
Collapse
|
49
|
Umeda T, Sakai A, Shigemori K, Yokota A, Kumagai T, Tomiyama T. Oligomer-Targeting Prevention of Neurodegenerative Dementia by Intranasal Rifampicin and Resveratrol Combination - A Preclinical Study in Model Mice. Front Neurosci 2021; 15:763476. [PMID: 34966254 PMCID: PMC8710719 DOI: 10.3389/fnins.2021.763476] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Amyloidogenic protein oligomers are thought to play an important role in the pathogenesis of neurodegenerative dementia, including Alzheimer’s disease, frontotemporal dementia, and dementia with Lewy bodies. Previously we demonstrated that oral or intranasal rifampicin improved the cognition of APP-, tau-, and α-synuclein-transgenic mice by reducing the amount of Aβ, tau, and α-synuclein oligomers in the brain. In the present study, to explore more effective and safer medications for dementia, we tested the drug combination of rifampicin and resveratrol, which is a multifunctional natural polyphenol with the potential to antagonize the adverse effects of rifampicin. The mixture was intranasally administered to APP-, tau-, and α-synuclein-transgenic mice, and their memory and oligomer-related pathologies were evaluated. Compared with rifampicin and resveratrol alone, the combinatorial medicine significantly improved mouse cognition, reduced amyloid oligomer accumulation, and recovered synaptophysin levels in the hippocampus. The plasma levels of liver enzymes, which reflect hepatic injury and normally increase by rifampicin treatment, remained normal by the combination treatment. Notably, resveratrol alone and the combinatorial medicine, but not rifampicin alone, enhanced the levels of brain-derived neurotrophic factor (BDNF) and its precursor, pro-BDNF, in the hippocampus. Furthermore, the combination showed a synergistic effect in ameliorating mouse cognition. These results show the advantages of this combinatorial medicine with regards to safety and effectiveness over single-drug rifampicin. Our findings may provide a feasible means for the prevention of neurodegenerative dementia that targets toxic oligomers.
Collapse
Affiliation(s)
- Tomohiro Umeda
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ayumi Sakai
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Keiko Shigemori
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ayumi Yokota
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | - Takami Tomiyama
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan.,Medilabo RFP, Inc., Kyoto, Japan
| |
Collapse
|
50
|
Zhai Y, Liu BG, Mo XN, Zou M, Mei XP, Chen W, Huang GD, Wu L. Gingerol ameliorates neuronal damage induced by hypoxia-reoxygenation via the miR-210/brain-derived neurotrophic factor axis. Kaohsiung J Med Sci 2021; 38:367-377. [PMID: 34962339 DOI: 10.1002/kjm2.12486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/22/2021] [Accepted: 10/15/2021] [Indexed: 01/15/2023] Open
Abstract
The specific mechanism of gingerol in cerebral ischemia remains unknown. A neuroprotective function for miR-210 in cerebral ischemia has been identified. The brain-derived neurotrophic factor (BDNF)-mediated signaling pathway protects against cerebral ischemic injury. This investigation aimed to determine whether gingerol plays a neuroprotective role in cerebral ischemia via the miR-210/BDNF axis. N2a cells subjected to 10 h of hypoxia and 4 h of reoxygenation were treated with 5, 10, or 20 μmol/L gingerol. The levels of viability, apoptosis, and proteins in N2a cells were determined using MTT assays, flow cytometry, and western blotting, respectively. The binding relationship between BDNF and miR-210 was studied using a dual luciferase reporter assay. The expression levels of miR-210 and BDNF were determined using qPCR. Gingerol repressed the increase in apoptosis and decrease in viability observed in response to hypoxia/reoxygenation. Gingerol increased Bcl-2, BDNF, and TrkB levels and reduced Bax and cleaved caspase 3 levels after hypoxia/reoxygenation. Gingerol evoked decreased expression of miR-210. Inhibition of miR-210 resulted in increased viability and reduced apoptosis along with increased levels of Bcl-2, BDNF, and TrkB and reduced levels of Bax and cleaved caspase 3 after hypoxia/reoxygenation. Additionally, the miR-210 mimic reversed changes induced by gingerol. The cotransfection of the miR-210 mimic and wild type BDNF led to decreased luciferase activity. BDNF was negatively regulated by miR-210. BDNF siRNA reversed these changes evoked by miR-210 inhibition. Gingerol ameliorated hypoxia/reoxygenation-stimulated neuronal damage by regulating the miR-210/BDNF axis, indicating that gingerol is worthy of further application in cerebral ischemia therapy.
Collapse
Affiliation(s)
- Yang Zhai
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China.,Department of Neurology, Guangxi Key Laboratory of Chinese Medicine Foundation Research, Nanning, China.,Department of International Medical, Guangxi International Zhuang Medicine Hospital, Nanning, China
| | - Bu-Gu Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Xue-Ni Mo
- Department of Neurology, Guangxi University of Chinese Medicine, Nanning, China
| | - Min Zou
- Department of Pediatrics, Guangxi International Zhuang Medicine Hospital, Nanning, China
| | - Xiao-Ping Mei
- Department of Endocrinology, Guangxi International Zhuang Medicine Hospital, Nanning, China
| | - Wei Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Guo-Dong Huang
- Department of International Medical, Guangxi International Zhuang Medicine Hospital, Nanning, China
| | - Lin Wu
- Department of Neurology, Guangxi Key Laboratory of Chinese Medicine Foundation Research, Nanning, China.,Department of Neurology, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|