1
|
Olawade DB, Rashad I, Egbon E, Teke J, Ovsepian SV, Boussios S. Reversing Epigenetic Dysregulation in Neurodegenerative Diseases: Mechanistic and Therapeutic Considerations. Int J Mol Sci 2025; 26:4929. [PMID: 40430067 PMCID: PMC12112518 DOI: 10.3390/ijms26104929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 05/05/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Epigenetic dysregulation has emerged as an important player in the pathobiology of neurodegenerative diseases (NDDs), such as Alzheimer's, Parkinson's, and Huntington's diseases. Aberrant DNA methylation, histone modifications, and dysregulated non-coding RNAs have been shown to contribute to neuronal dysfunction and degeneration. These alterations are often exacerbated by environmental toxins, which induce oxidative stress, inflammation, and genomic instability. Reversing epigenetic aberrations may offer an avenue for restoring brain mechanisms and mitigating neurodegeneration. Herein, we revisit the evidence suggesting the ameliorative effects of epigenetic modulators in toxin-induced models of NDDs. The restoration of normal gene expressions, the improvement of neuronal function, and the reduction in pathological markers by histone deacetylase (HDAC) and DNA methyltransferase (DNMT) inhibitors have been demonstrated in preclinical models of NDDs. Encouragingly, in clinical trials of Alzheimer's disease (AD), HDAC inhibitors have caused improvements in cognition and memory. Combining these beneficial effects of epigenetic modulators with neuroprotective agents and the clearance of misfolded amyloid proteins may offer synergistic benefits. Reinforced by the emerging methods for more effective and brain-specific delivery, reversibility, and safety considerations, epigenetic modulators are anticipated to minimize systemic toxicity and yield more favorable outcomes in NDDs. In summary, although still in their infancy, epigenetic modulators offer an integrated strategy to address the multifactorial nature of NDDs, altering their therapeutic landscape.
Collapse
Affiliation(s)
- David B. Olawade
- Department of Allied and Public Health, School of Health, Sport and Bioscience, University of East London, London E16 2RD, UK;
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK;
- Department of Public Health, York St John University, London E14 2BA, UK
- School of Health and Care Management, Arden University, Arden House, Middlemarch Park, Coventry CV3 4FJ, UK
| | - Intishar Rashad
- Department of Acute Medicine, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK
| | - Eghosasere Egbon
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Life Science Engineering, FH Technikum, 1200 Vienna, Austria;
| | - Jennifer Teke
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK;
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury CT1 1QU, UK
| | - Saak Victor Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime ME4 4TB, UK;
- Faculty of Medicine, Tbilisi State University, Tbilisi 0177, Georgia
| | - Stergios Boussios
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK;
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury CT1 1QU, UK
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK
| |
Collapse
|
2
|
A E F Cardinali C, Martins YA, C M Moraes R, Costa AP, Torrão AS. Benfotiamine Ameliorates Streptozotocin-Induced Alzheimer's Disease in Rats by Modulating Neuroinflammation, Oxidative Stress, and Microglia. Mol Neurobiol 2025:10.1007/s12035-025-04811-x. [PMID: 40038195 DOI: 10.1007/s12035-025-04811-x] [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: 10/14/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia, characterized by progressive memory loss and cognitive decline. Recent evidence indicates that inflammation plays a central role in AD pathogenesis, with elevated inflammatory markers and risk genes linked to innate immune functions. Glial cell dysfunction, particularly in astrocytes and microglia, is crucial to the neuroinflammatory process, contributing to oxidative stress, synaptic dysfunction, neuronal death, and impaired neurogenesis. This study aimed to investigate the therapeutic effects of benfotiamine (BFT), a vitamin B1 analogue, on microglial morphology, inflammation, and oxidative stress parameters in a sporadic Alzheimer-like disease model induced by intracerebroventricular injection of streptozotocin (STZ). Supplementation with 150 mg/kg of BFT for 7 days significantly reduced inflammation in the hippocampus and provided protection against oxidative damage in the entorhinal cortex by activating the Nrf-2 pathway and enhancing the expression of antioxidant enzymes such as SOD1 and CAT. These findings suggest that BFT exerts neuroprotective effects in AD, particularly impacting glial cell function and redox homeostasis.
Collapse
Affiliation(s)
- Camila A E F Cardinali
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil.
| | - Yandara A Martins
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| | - Ruan C M Moraes
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
- Department of Psychiatry & Behavioral Neurobiology, The University of Alabama at Birmingham, Alabama, USA
| | - Andressa P Costa
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| | - Andréa S Torrão
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| |
Collapse
|
3
|
Krishnamurthy HK, Jayaraman V, Krishna K, Wang T, Bei K, Changalath C, Rajasekaran JJ. An overview of the genes and biomarkers in Alzheimer's disease. Ageing Res Rev 2025; 104:102599. [PMID: 39612989 DOI: 10.1016/j.arr.2024.102599] [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: 09/03/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and neurodegenerative disease characterized by neurofibrillary tangles (NFTs) and amyloid plaque. Familial AD is caused by mutations in the APP, PSEN1, and PSEN2 genes and these mutations result in the early onset of the disease. Sporadic AD usually affects older adults over the age of 65 years and is, therefore classified as late-onset AD (LOAD). Several risk factors associated with LOAD including the APOE gene have been identified. Moreover, GWAS studies have identified a wide array of genes and polymorphisms that are associated with LOAD risk. Currently, the diagnosis of AD involves the evaluation of memory and personality changes, cognitive impairment, and medical and family history to rule out other diseases. Laboratory tests to assess the biomarkers in the body fluids as well as MRI, CT, and PET scans to analyze the presence of plaques and NFTs are also included in the diagnosis of AD. It is important to diagnose AD before the onset of clinical symptoms, i.e. during the preclinical stage, to delay the progression and for better management of the disease. Research has been conducted to identify biomarkers of AD in the CSF, serum, saliva, and urine during the preclinical stage. Current research has identified several biomarkers and potential biomarkers in the body fluids that enhance diagnostic accuracy. Aside from genetics, other factors such as diet, physical activity, and lifestyle factors may influence the risk of developing AD. Clinical trials are underway to find potential biomarkers, diagnostic measures, and treatments for AD mainly in the preclinical stage. This review provides an overview of the genes and biomarkers of AD.
Collapse
Affiliation(s)
| | | | - Karthik Krishna
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Tianhao Wang
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Kang Bei
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | | | | |
Collapse
|
4
|
Hata T, Grenier F, Hiraga T, Soya M, Okamoto M, Soya H. Promoting arousal associated with physical activity with the vitamin B 1 derivative TTFD. J Physiol Sci 2024; 75:100001. [PMID: 40008856 PMCID: PMC11979667 DOI: 10.1016/j.jphyss.2024.100001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 02/27/2025]
Abstract
Physical inactivity, which is a global issue, reduces physical and mental vitality, particularly impairing prefrontal-cortex-based mental health. This may trigger social withdrawal and depression, hindering the ability to have an active lifestyle. However, we have identified a beneficial agent, a vitamin B1 derivative called thiamine tetrahydrofurfuryl disulfide (TTFD), that enhances physical activity through dopaminergic regulation in the medial prefrontal cortex (mPFC) of rats. Since the brain dopaminergic system also regulates the sleep-wake cycle via the ascending arousal system, we postulated that TTFD may promote arousal. To test this, we performed electroencephalograms and electromyograms in rats, monitoring their physical activity and sleep-wake cycles after TTFD injection. Analysis revealed that TTFD acutely promotes arousal, reduces slow-wave sleep (SWS) and rapid eye movement (REM) sleep, and promotes increased physical activity. TTFD not only promotes physical activity but also increases arousal, thereby potentially contributing to enhanced mental health.
Collapse
Affiliation(s)
- Toshiaki Hata
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan; Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan
| | - François Grenier
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan
| | - Taichi Hiraga
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan
| | - Mariko Soya
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan
| | - Masahiro Okamoto
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan; Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan; Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan.
| |
Collapse
|
5
|
Preethy H A, Rajendran K, Sukumar AJ, Krishnan UM. Emerging paradigms in Alzheimer's therapy. Eur J Pharmacol 2024; 981:176872. [PMID: 39117266 DOI: 10.1016/j.ejphar.2024.176872] [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: 03/08/2024] [Revised: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Alzheimer's disease is a neurodegenerative disorder that affects elderly, and its incidence is continuously increasing across the globe. Unfortunately, despite decades of research, a complete cure for Alzheimer's disease continues to elude us. The current medications are mainly symptomatic and slow the disease progression but do not result in reversal of all disease pathologies. The growing body of knowledge on the factors responsible for the onset and progression of the disease has resulted in the identification of new targets that could be targeted for treatment of Alzheimer's disease. This has opened new vistas for treatment of Alzheimer's disease that have moved away from chemotherapeutic agents modulating a single target to biologics and combinations that acted on multiple targets thereby offering better therapeutic outcomes. This review discusses the emerging directions in therapeutic interventions against Alzheimer's disease highlighting their merits that promise to change the treatment paradigm and challenges that limit their clinical translation.
Collapse
Affiliation(s)
- Agnes Preethy H
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Kayalvizhi Rajendran
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Anitha Josephine Sukumar
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Uma Maheswari Krishnan
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India; School of Arts, Sciences, Humanities & Education, SASTRA Deemed University, Thanjavur, India.
| |
Collapse
|
6
|
Zhang L, Luo YL, Xiang Y, Bai XY, Qiang RR, Zhang X, Yang YL, Liu XL. Ferroptosis inhibitors: past, present and future. Front Pharmacol 2024; 15:1407335. [PMID: 38846099 PMCID: PMC11153831 DOI: 10.3389/fphar.2024.1407335] [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: 03/26/2024] [Accepted: 05/06/2024] [Indexed: 06/09/2024] Open
Abstract
Ferroptosis is a non-apoptotic mode of programmed cell death characterized by iron dependence and lipid peroxidation. Since the ferroptosis was proposed, researchers have revealed the mechanisms of its formation and continue to explore effective inhibitors of ferroptosis in disease. Recent studies have shown a correlation between ferroptosis and the pathological mechanisms of neurodegenerative diseases, as well as diseases involving tissue or organ damage. Acting on ferroptosis-related targets may provide new strategies for the treatment of ferroptosis-mediated diseases. This article specifically describes the metabolic pathways of ferroptosis and summarizes the reported mechanisms of action of natural and synthetic small molecule inhibitors of ferroptosis and their efficacy in disease. The paper also describes ferroptosis treatments such as gene therapy, cell therapy, and nanotechnology, and summarises the challenges encountered in the clinical translation of ferroptosis inhibitors. Finally, the relationship between ferroptosis and other modes of cell death is discussed, hopefully paving the way for future drug design and discovery.
Collapse
Affiliation(s)
- Lei Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yi Lin Luo
- School of Medicine, Yan’an University, Yan’an, China
| | - Yang Xiang
- College of Physical Education, Yan’an University, Yan’an, China
| | - Xin Yue Bai
- School of Medicine, Yan’an University, Yan’an, China
| | | | - Xin Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yan Ling Yang
- School of Medicine, Yan’an University, Yan’an, China
| | - Xiao Long Liu
- School of Medicine, Yan’an University, Yan’an, China
| |
Collapse
|
7
|
Bozic I, Lavrnja I. Thiamine and benfotiamine: Focus on their therapeutic potential. Heliyon 2023; 9:e21839. [PMID: 38034619 PMCID: PMC10682628 DOI: 10.1016/j.heliyon.2023.e21839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Thiamine, also known as vitamin B1, is an essential nutrient that plays a crucial role in energy metabolism and overall health. It is a water-soluble vitamin that plays an important role in the conversion of carbohydrates into energy in the body. Thiamine is essential for the proper functioning of the nervous system, heart and muscles. Thiamine deficiency is a life-threatening disease that leads to various disorders and lesions in the nerves and brain, at least in vertebrates. Several thiamine precursors with higher bioavailability have been developed to compensate for thiamine deficiency, including benfotiamine. Benfotiamine is more bioavailable and has higher tissue penetration than thiamine. Studies have shown its antioxidant and anti-inflammatory potential in activated immune and glial cells. It also improves complications observed in type 2 diabetes and has beneficial effects in mouse models of neurodegenerative disease. Benfotiamine represents an off-the-shelf agent used to support nerve health, promote healthy aging and support glucose metabolism. Accordingly, the present review aimed to provide an overview of the neuroprotective effects of thiamine/benfotiamine in the context of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Iva Bozic
- Institute for Biological Research "Sinisa Stankovic"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
8
|
Bettendorff L. Synthetic Thioesters of Thiamine: Promising Tools for Slowing Progression of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:11296. [PMID: 37511056 PMCID: PMC10379298 DOI: 10.3390/ijms241411296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Thiamine (vitamin B1) is essential for the brain. This is attributed to the coenzyme role of thiamine diphosphate (ThDP) in glucose and energy metabolism. The synthetic thiamine prodrug, the thioester benfotiamine (BFT), has been extensively studied and has beneficial effects both in rodent models of neurodegeneration and in human clinical studies. BFT has no known adverse effects and improves cognitive outcomes in patients with mild Alzheimer's disease. In cell culture and animal models, BFT has antioxidant and anti-inflammatory properties that seem to be mediated by a mechanism independent of the coenzyme function of ThDP. Recent in vitro studies show that another thiamine thioester, O,S-dibenzoylthiamine (DBT), is even more efficient than BFT, especially with respect to its anti-inflammatory potency, and is effective at lower concentrations. Thiamine thioesters have pleiotropic properties linked to an increase in circulating thiamine concentrations and possibly in hitherto unidentified open thiazole ring derivatives. The identification of the active neuroprotective metabolites and the clarification of their mechanism of action open extremely promising perspectives in the field of neurodegenerative, neurodevelopmental, and psychiatric conditions. The present review aims to summarize existing data on the neuroprotective effects of thiamine thioesters and give a comprehensive account.
Collapse
Affiliation(s)
- Lucien Bettendorff
- Laboratory of Neurophysiology, GIGA Neurosciences, University of Liège, 4000 Liège, Belgium
| |
Collapse
|
9
|
Mann RH. Impaired Thiamine Metabolism in Amyotrophic Lateral Sclerosis and Its Potential Treatment With Benfotiamine: A Case Report and a Review of the Literature. Cureus 2023; 15:e40511. [PMID: 37333039 PMCID: PMC10274516 DOI: 10.7759/cureus.40511] [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] [Accepted: 06/15/2023] [Indexed: 06/20/2023] Open
Abstract
Homogenates of brain tissue from the frontal cortex at autopsy in patients with amyotrophic lateral sclerosis (ALS) showed dramatically reduced levels of the enzyme thiamine pyrophosphatase (TPPase), the enzyme responsible for the conversion of thiamine pyrophosphate (TPP) to thiamine monophosphate (TMP). Additionally, free thiamine (vitamin B1) and TMP levels have been shown to be significantly reduced in the plasma and cerebral spinal fluid (CSF) of patients with ALS. These findings suggest that there is impaired thiamine metabolism in patients with ALS. Impaired thiamine metabolism decreases adenosine triphosphate (ATP) production and is a well-established cause of neurodegeneration. Decreased levels of TPPase, resulting in decreased levels of TMP in the cells of the frontal cortex, might account for the focal neurodegenerative changes observed in motor neurons in ALS. Benfotiamine, a safe, lipid-soluble, highly absorbable thiamine analogue, significantly raises free thiamine, TMP, and TPP levels in the blood. A case in which benfotiamine may have positively impacted the symptoms of a patient with ALS is presented. The use of benfotiamine in patients with ALS appears to be a promising therapeutic option. Considering the severity and the lack of satisfactory treatment options associated with this disease, more research on the effects of benfotiamine on the course of ALS is urgently needed.
Collapse
|
10
|
Revisiting the Role of Vitamins and Minerals in Alzheimer's Disease. Antioxidants (Basel) 2023; 12:antiox12020415. [PMID: 36829974 PMCID: PMC9952129 DOI: 10.3390/antiox12020415] [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: 01/16/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia that affects millions of individuals worldwide. It is an irreversible neurodegenerative disorder that is characterized by memory loss, impaired learning and thinking, and difficulty in performing regular daily activities. Despite nearly two decades of collective efforts to develop novel medications that can prevent or halt the disease progression, we remain faced with only a few options with limited effectiveness. There has been a recent growth of interest in the role of nutrition in brain health as we begin to gain a better understanding of what and how nutrients affect hormonal and neural actions that not only can lead to typical cardiovascular or metabolic diseases but also an array of neurological and psychiatric disorders. Vitamins and minerals, also known as micronutrients, are elements that are indispensable for functions including nutrient metabolism, immune surveillance, cell development, neurotransmission, and antioxidant and anti-inflammatory properties. In this review, we provide an overview on some of the most common vitamins and minerals and discuss what current studies have revealed on the link between these essential micronutrients and cognitive performance or AD.
Collapse
|
11
|
Blaylock RL. The biochemical basis of neurodegenerative disease: The role of immunoexcitoxicity and ways to possibly attenuate it. Surg Neurol Int 2023; 14:141. [PMID: 37151454 PMCID: PMC10159298 DOI: 10.25259/sni_250_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 05/09/2023] Open
Abstract
There is growing evidence that inflammation secondary to immune activation is intimately connected to excitotoxicity. We now know that most peripheral tissues contain fully operational glutamate receptors. While most of the available research deals with excitotoxicity in central nervous system (CNS) tissues, this is no longer true. Even plant has been found to contain glutamate receptors. Most of the immune cells, including mask cells, contain glutamate receptors. The receptors are altered by inflammation, both chemokine and cytokines. A host of new diseases have been found that are caused by immunity to certain glutamate receptors, as we see with Rasmussen's encephalitis. In this paper, I try to explain this connection and possible ways to reduce or even stop the reaction.
Collapse
Affiliation(s)
- Russell L. Blaylock
- Corresponding author: Russell L. Blaylock, M.D. 609 Old Natchez Trace Canton, MS. Retired Neurosurgeon, Department of Neurosurgery, Theoretical Neuroscience Research, LLC, Ridgeland, Mississippi, United States.
| |
Collapse
|
12
|
Zhang Y, Wang M, Chang W. Iron dyshomeostasis and ferroptosis in Alzheimer’s disease: Molecular mechanisms of cell death and novel therapeutic drugs and targets for AD. Front Pharmacol 2022; 13:983623. [PMID: 36188557 PMCID: PMC9523169 DOI: 10.3389/fphar.2022.983623] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a degenerative disease of the central nervous system that is the most common type of senile dementia. Ferroptosis is a new type of iron-dependent programmed cell death identified in recent years that is different from other cell death forms. Ferroptosis is induced by excessive accumulation of lipid peroxides and reactive oxygen species (ROS) in cells. In recent years, it has been found that ferroptosis plays an important role in the pathological process of AD. Iron dyshomeostasis contribute to senile plaques (SP) deposition and neurofibrillary tangles (NFTs). Iron metabolism imbalance in brain and the dysfunction of endogenous antioxidant systems including system Xc- and glutathione peroxidase (GPX) are closely related to the etiopathogenesis of AD. Dysfunction of nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy induced ferroptosis can accelerates the pathological process of AD. In addition, NRF2, through regulating the expression of a considerable number of genes related to ferroptosis, including genes related to iron and glutathione metabolism, plays an important role in the development of AD. Here, we review the potential interaction between AD and ferroptosis and the major pathways regulating ferroptosis in AD. We also review the active natural and synthetic compounds such as iron chelators, lipid peroxidation inhibitors and antioxidants available to treat AD by alleviating iron dyshomeostasis and preventing ferroptosis in mice and cell models to provide valuable information for the future treatment and prevention of AD.
Collapse
|
13
|
de Moraes RCM, Lima GCA, Cardinali CAEF, Gonçalves AC, Portari GV, Guerra-Shinohara EM, Leboucher A, Júnior JD, Kleinridders A, da Silva Torrão A. Benfotiamine protects against hypothalamic dysfunction in a STZ-induced model of neurodegeneration in rats. Life Sci 2022; 306:120841. [PMID: 35907494 DOI: 10.1016/j.lfs.2022.120841] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/13/2022] [Accepted: 07/22/2022] [Indexed: 10/16/2022]
Abstract
The neurodegeneration of Alzheimer's disease (AD) affects not only brain structures associate with cognition early in the progression of the disease, but other areas such as the hypothalamus, a region involved in the control of metabolism and appetite. In this context, we evaluated the effects of benfotiamine (BFT), a vitamin B1 analog that is being proposed as a therapeutical approach for AD-related cognitive alterations, which were induced by intracerebroventricular injection of streptozotocin (STZ). In addition to the already described effect of STZ on cognition, we show that this drug also causes metabolic changes which are linked to changes in hypothalamic insulin signaling and orexigenic and anorexigenic circuitries, as well as a decreased cellular integrated stress response. As expected, the supplementation with 150 mg/kg of BFT for 30 days increased blood concentrations of thiamine and its phosphate esters. This led to the prevention of body weight and fat loss in STZ-ICV-treated animals. In addition, we also found an improvement in food consumption, despite hypothalamic gene expression linked to anorexia after STZ exposure. Additionally, decreased apoptosis signaling was observed in the hypothalamus. In in vitro experiments, we noticed a high ability of BFT to increase insulin sensitivity in hypothalamic neurons. Furthermore, we also observed that BFT decreases the mitochondrial unfolded stress response damage by preventing the loss of HSP60 and reversed the mitochondria dysfunction caused by STZ. Taken together, these results suggest that benfotiamine treatment is a potential therapeutic approach in the treatment of hypothalamic dysfunction and metabolic disturbances associated with sporadic AD.
Collapse
Affiliation(s)
- Ruan Carlos Macêdo de Moraes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Brazil; Central Regulation of Metabolism, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany.
| | | | | | - Alisson Carvalho Gonçalves
- Federal Institute of Education, Science and Technology Goiano, Urutaí, GO, Brazil; Laboratory of Experimental Nutrition, Institute of Health Sciences, Federal University of Triângulo Mineiro, Brazil
| | - Guilherme Vannucchi Portari
- Laboratory of Experimental Nutrition, Institute of Health Sciences, Federal University of Triângulo Mineiro, Brazil
| | - Elvira Maria Guerra-Shinohara
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Brazil; Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul, Brazil
| | - Antoine Leboucher
- Central Regulation of Metabolism, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
| | - José Donato Júnior
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | - André Kleinridders
- Central Regulation of Metabolism, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany; Institute of Nutritional Science, Department of Molecular and Experimental Nutritional Medicine, University of Potsdam, Germany
| | - Andréa da Silva Torrão
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Brazil
| |
Collapse
|
14
|
Therapeutic potential of vitamin B 1 derivative benfotiamine from diabetes to COVID-19. Future Med Chem 2022; 14:809-826. [PMID: 35535731 DOI: 10.4155/fmc-2022-0040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Benfotiamine (S-benzoylthiamine-O-monophosphate), a unique, lipid-soluble derivative of thiamine, is the most potent allithiamine found in roasted garlic, as well as in other herbs of the genus Allium. In addition to potent antioxidative properties, benfotiamine has also been shown to be a strong anti-inflammatory agent with therapeutic significance to several pathological complications. Specifically, over the past decade or so, benfotiamine has been shown to prevent not only various secondary diabetic complications but also several inflammatory complications such as uveitis and endotoxemia. Recent studies also demonstrate that this compound could be used to prevent the symptoms associated with various infectious diseases such as HIV and COVID-19. In this review article, the authors discuss the significance of benfotiamine in the prevention of various pathological complications.
Collapse
|
15
|
Qian T, Zhao L, Pan X, Sang S, Xu Y, Wang C, Zhong C, Fei G, Cheng X. Association Between Blood Biochemical Factors Contributing to Cognitive Decline and B Vitamins in Patients With Alzheimer's Disease. Front Nutr 2022; 9:823573. [PMID: 35265656 PMCID: PMC8898888 DOI: 10.3389/fnut.2022.823573] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/27/2022] [Indexed: 01/01/2023] Open
Abstract
Background Malnutrition, metabolism stress, inflammation, peripheral organs dysfunction, and B vitamins deficiency significantly contribute to the progression and mortality of Alzheimer's disease (AD). However, it is unclear which blood biochemical indicators are most closely related to cognitive decline and B vitamins deficiency (thiamine, folate, vitamin B12) in patients with AD. Methods This was a cross-sectional study of 206 AD patients recruited from six hospitals in China. Thiamine diphosphate (TDP), the bioactive form of thiamine, was measured by high-performance liquid chromatography fluoroscopy (HPLC) at a single center. Levels of biochemical indicators (except TDP) were measured by regular and standard laboratory tests in each hospital. Pearson's rank correlation analysis was used to assess relationships between B vitamins and biochemical indicators. T-test was used to compare the difference between ApoE ε4 and non-ApoE ε4 groups. Differences were considered statistically significant as P < 0.05. Results Among the biochemical results, in AD population, malnutrition indicators (erythrocyte, hemoglobin, serum albumin, and total protein) were most significantly associated with cognitive function, as was free triiodothyronine (FT3) levels which had been observed in previous study. Malnutrition and FT3 levels depend on age but not apolipoprotein E (ApoE) genotype. Meanwhile, Among the B vitamins, TDP was the most significantly associated with malnutrition indicators and FT3. Conclusion Our results indicated that TDP reduction could be a modifiable risk factor for malnutrition and FT3 that contributed to cognitive decline in AD patients. Correcting thiamine metabolism could serve as an optional therapy target for AD treatment.
Collapse
Affiliation(s)
- Ting Qian
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Lei Zhao
- Department of Neurology, Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoli Pan
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Shaoming Sang
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yangqi Xu
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Changpeng Wang
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Guoqiang Fei
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Xiaoqin Cheng
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Increasing Inhibition of the Rat Brain 2-Oxoglutarate Dehydrogenase Decreases Glutathione Redox State, Elevating Anxiety and Perturbing Stress Adaptation. Pharmaceuticals (Basel) 2022; 15:ph15020182. [PMID: 35215295 PMCID: PMC8875720 DOI: 10.3390/ph15020182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/10/2022] Open
Abstract
Specific inhibitors of mitochondrial 2-oxoglutarate dehydrogenase (OGDH) are administered to animals to model the downregulation of the enzyme as observed in neurodegenerative diseases. Comparison of the effects of succinyl phosphonate (SP, 0.02 mmol/kg) and its uncharged precursor, triethyl succinyl phosphonate (TESP, 0.02 and 0.1 mmol/kg) reveals a biphasic response of the rat brain metabolism and physiology to increasing perturbation of OGDH function. At the low (TE)SP dose, glutamate, NAD+, and the activities of dehydrogenases of 2-oxoglutarate and malate increase, followed by their decreases at the high TESP dose. The complementary changes, i.e., an initial decrease followed by growth, are demonstrated by activities of pyruvate dehydrogenase and glutamine synthetase, and levels of oxidized glutathione and citrulline. While most of these indicators return to control levels at the high TESP dose, OGDH activity decreases and oxidized glutathione increases, compared to their control values. The first phase of metabolic perturbations does not cause significant physiological changes, but in the second phase, the ECG parameters and behavior reveal decreased adaptability and increased anxiety. Thus, lower levels of OGDH inhibition are compensated by the rearranged metabolic network, while the increased levels induce a metabolic switch to a lower redox state of the brain, associated with elevated stress of the animals.
Collapse
|
17
|
Vitamin B1, eye and brain. SRP ARK CELOK LEK 2022. [DOI: 10.2298/sarh210929019b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Vitamin B1 (aneurin, thiamine) is a water-soluble vitamin necessary for the
normal function of the nervous system, visual system and heart and is part
of important enzymes in the body. Thiamine enables the normal use of
glucose, other carbohydrates and proteins, and enables the supply of energy
to the organism. The main sources of thiamine are exogenous and small
amounts are synthesized by microorganisms of the human intestinal
microbiome. Vitamin B1 cannot accumulate in the body, so signs of
deficiency are quickly manifested. Hypovitaminosis B1 is seen in chronic
ethyl abuse, persistent vomiting (as in some pregnant women) or after
bariatric surgical procedures, but in a mild form it is present in the
general population. Normal daily needs for vitamin B1 depend on calorie
intake, and 0.4 mg should be ingested for every 1000 kcal.
Collapse
|
18
|
Sambon M, Wins P, Bettendorff L. Neuroprotective Effects of Thiamine and Precursors with Higher Bioavailability: Focus on Benfotiamine and Dibenzoylthiamine. Int J Mol Sci 2021; 22:ijms22115418. [PMID: 34063830 PMCID: PMC8196556 DOI: 10.3390/ijms22115418] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 11/25/2022] Open
Abstract
Thiamine (vitamin B1) is essential for brain function because of the coenzyme role of thiamine diphosphate (ThDP) in glucose and energy metabolism. In order to compensate thiamine deficiency, several thiamine precursors with higher bioavailability were developed since the 1950s. Among these, the thioester benfotiamine (BFT) has been extensively studied and has beneficial effects both in rodent models of neurodegeneration and in human clinical studies. BFT has antioxidant and anti-inflammatory properties that seem to be mediated by a mechanism independent of the coenzyme function of ThDP. BFT has no adverse effects and improves cognitive outcome in patients with mild Alzheimer’s disease (AD). Recent in vitro studies show that another thiamine thioester, dibenzoylthiamine (DBT) is even more efficient that BFT, especially with respect to its anti-inflammatory potency. Thiamine thioesters have pleiotropic properties linked to an increase in circulating thiamine concentrations and possibly in hitherto unidentified metabolites in particular open thiazole ring derivatives. The identification of the active neuroprotective derivatives and the clarification of their mechanism of action open extremely promising perspectives in the field of neurodegenerative, neurodevelopmental and psychiatric conditions.
Collapse
|
19
|
Brain Energy Improvement as an Emerging Approach for Alzheimer's Disease Treatment. Neurosci Bull 2021; 37:892-893. [PMID: 33861413 DOI: 10.1007/s12264-021-00679-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/09/2021] [Indexed: 10/21/2022] Open
|
20
|
Sheng L, Cao W, Lin P, Chen W, Xu H, Zhong C, Yuan F, Chen H, Li H, Liu C, Yang M, Li X. Safety, Tolerability and Pharmacokinetics of Single and Multiple Ascending Doses of Benfotiamine in Healthy Subjects. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:1101-1110. [PMID: 33727798 PMCID: PMC7955752 DOI: 10.2147/dddt.s296197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
Purpose Safety, tolerability and pharmacokinetics of single and multiple ascending doses (SADs/MADs) of benfotiamine were assessed after oral administration in two randomized, double-blind, placebo-controlled, phase I trials. Methods Healthy subjects were sequentially enrolled into one of five SAD (150–1200 mg) or three MAD (150, 300 or 600 mg) cohorts. In SAD study, each cohort of 12 subjects (n = 10, active; n = 2, placebo) were administrated once-daily doses. In MAD study, each cohort of 16 subjects (n = 12, active; n = 4, placebo) were administrated once-daily on day 1 and twice-daily on day 4–9, followed by a single morning dose on day 10. Results In the SAD study, the median time to reach maximum concentration (Tmax) arrived 1.0 to 2.0 h for thiamine (TM), 3.5 to 8.0 h for thiamine monophosphate (TMP), and 8.0 to 24.0 h for thiamine diphosphate (TDP) after administration of benfotiamine. The area under concentration-time curve from 0 to last measurable concentration (AUC0-t) or maximum observed concentration (Cmax) of TM, TMP, and TDP was less or more dose proportional over the single dose studied except Cmax of TM. Food consumption did not increase the level of TM and TDP at baseline. TM exhibited a relatively long elimination half-life (t1/2) in all doses studied, resulting in accumulation ratio (Rac) of 1.96 to 2.11 and accumulation ratio based on Cmax (Rac, Cmax) of 1.60 to 1.88 following 7 days of multiple dosing. Comparable accumulation results were also obtained for TDP after multiple dosing. The incidence and severity of adverse events (AEs) were similar between benfotiamine and placebo. The commonly reported drug-related AEs were increased ALT and urinary WBC. Conclusion Both SAD and MAD studies of benfotiamine in healthy subjects were safe and well tolerated. TM and TDP exhibited moderate accumulation on repeated administration of benfotiamine.
Collapse
Affiliation(s)
- Lei Sheng
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wei Cao
- Shanghai Rixin Biotechnology Co., Ltd, Shanghai, 200237, People's Republic of China
| | - Pingping Lin
- Phase I Clinical Research Center, The Affiliated Hospital of Qingdao University, Shandong, 266071, People's Republic of China
| | - Weili Chen
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Hongrong Xu
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Fei Yuan
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Hanjing Chen
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Hui Li
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chao Liu
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Mengjie Yang
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xuening Li
- Department of Clinical Pharmacology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| |
Collapse
|
21
|
Sambon M, Gorlova A, Demelenne A, Alhama-Riba J, Coumans B, Lakaye B, Wins P, Fillet M, Anthony DC, Strekalova T, Bettendorff L. Dibenzoylthiamine Has Powerful Antioxidant and Anti-Inflammatory Properties in Cultured Cells and in Mouse Models of Stress and Neurodegeneration. Biomedicines 2020; 8:biomedicines8090361. [PMID: 32962139 PMCID: PMC7555733 DOI: 10.3390/biomedicines8090361] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/03/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
Thiamine precursors, the most studied being benfotiamine (BFT), have protective effects in mouse models of neurodegenerative diseases. BFT decreased oxidative stress and inflammation, two major characteristics of neurodegenerative diseases, in a neuroblastoma cell line (Neuro2a) and an immortalized brain microglial cell line (BV2). Here, we tested the potential antioxidant and anti-inflammatory effects of the hitherto unexplored derivative O,S-dibenzoylthiamine (DBT) in these two cell lines. We show that DBT protects Neuro2a cells against paraquat (PQ) toxicity by counteracting oxidative stress at low concentrations and increases the synthesis of reduced glutathione and NADPH in a Nrf2-independent manner. In BV2 cells activated by lipopolysaccharides (LPS), DBT significantly decreased inflammation by suppressing translocation of NF-κB to the nucleus. Our results also demonstrate the superiority of DBT over thiamine and other thiamine precursors, including BFT, in all of the in vitro models. Finally, we show that the chronic administration of DBT arrested motor dysfunction in FUS transgenic mice, a model of amyotrophic lateral sclerosis, and it reduced depressive-like behavior in a mouse model of ultrasound-induced stress in which it normalized oxidative stress marker levels in the brain. Together, our data suggest that DBT may have therapeutic potential for brain pathology associated with oxidative stress and inflammation by novel, coenzyme-independent mechanisms.
Collapse
Affiliation(s)
- Margaux Sambon
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, 4000 Liège, Belgium; (M.S.); (J.A.-R.); (P.W.)
| | - Anna Gorlova
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands; (A.G.); (T.S.)
- Institute of Molecular Medicine Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Alice Demelenne
- Laboratory for the Analysis of Medicines, CIRM, Department of Pharmacy, University of Liège, 4000 Liège, Belgium; (A.D.); (M.F.)
| | - Judit Alhama-Riba
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, 4000 Liège, Belgium; (M.S.); (J.A.-R.); (P.W.)
- Faculty of Sciences, University of Girona, 17004 Girona, Spain
| | - Bernard Coumans
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Stem Cell, University of Liège, 4000 Liège, Belgium; (B.C.); (B.L.)
| | - Bernard Lakaye
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Stem Cell, University of Liège, 4000 Liège, Belgium; (B.C.); (B.L.)
| | - Pierre Wins
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, 4000 Liège, Belgium; (M.S.); (J.A.-R.); (P.W.)
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines, CIRM, Department of Pharmacy, University of Liège, 4000 Liège, Belgium; (A.D.); (M.F.)
| | - Daniel C. Anthony
- Institute of Molecular Medicine Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Department of Pharmacology, Oxford University, Oxford OX1 3QT, UK
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands; (A.G.); (T.S.)
- Institute of Molecular Medicine Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Lucien Bettendorff
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, 4000 Liège, Belgium; (M.S.); (J.A.-R.); (P.W.)
- Correspondence: ; Tel.: +32-4-366-5967
| |
Collapse
|
22
|
Jain R, Özgümüş T, Jensen TM, du Plessis E, Keindl M, Møller CL, Falhammar H, Nyström T, Catrina SB, Jörneskog G, Jessen LE, Forsblom C, Haukka JK, Groop PH, Rossing P, Groop L, Eliasson M, Eliasson B, Brismar K, Al-Majdoub M, Nilsson PM, Taskinen MR, Ferrannini E, Spégel P, Berg TJ, Lyssenko V. Liver nucleotide biosynthesis is linked to protection from vascular complications in individuals with long-term type 1 diabetes. Sci Rep 2020; 10:11561. [PMID: 32665614 PMCID: PMC7360755 DOI: 10.1038/s41598-020-68130-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Identification of biomarkers associated with protection from developing diabetic complications is a prerequisite for an effective prevention and treatment. The aim of the present study was to identify clinical and plasma metabolite markers associated with freedom from vascular complications in people with very long duration of type 1 diabetes (T1D). Individuals with T1D, who despite having longer than 30 years of diabetes duration never developed major macro- or microvascular complications (non-progressors; NP) were compared with those who developed vascular complications within 25 years from diabetes onset (rapid progressors; RP) in the Scandinavian PROLONG (n = 385) and DIALONG (n = 71) cohorts. The DIALONG study also included 75 healthy controls. Plasma metabolites were measured using gas and/or liquid chromatography coupled to mass spectrometry. Lower hepatic fatty liver indices were significant common feature characterized NPs in both studies. Higher insulin sensitivity and residual ß-cell function (C-peptide) were also associated with NPs in PROLONG. Protection from diabetic complications was associated with lower levels of the glycolytic metabolite pyruvate and APOCIII in PROLONG, and with lower levels of thiamine monophosphate and erythritol, a cofactor and intermediate product in the pentose phosphate pathway as well as higher phenylalanine, glycine and serine in DIALONG. Furthermore, T1D individuals showed elevated levels of picolinic acid as compared to the healthy individuals. The present findings suggest a potential beneficial shunting of glycolytic substrates towards the pentose phosphate and one carbon metabolism pathways to promote nucleotide biosynthesis in the liver. These processes might be linked to higher insulin sensitivity and lower liver fat content, and might represent a mechanism for protection from vascular complications in individuals with long-term T1D.
Collapse
Affiliation(s)
- Ruchi Jain
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden
| | - Türküler Özgümüş
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, 5032, Bergen, Norway
| | - Troels Mygind Jensen
- Research Unit for General Practice, Danish Aging Research Center, University of Southern Denmark, Odense, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Elsa du Plessis
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, 5032, Bergen, Norway
| | - Magdalena Keindl
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, 5032, Bergen, Norway
| | | | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Division of Internal Medicine, Unit for Diabetes Research, Karolinska Institute, South Hospital, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Center for Diabetes, Academica Specialist Centrum, Stockholm, Sweden
| | - Gun Jörneskog
- Department of Clinical Sciences, Division of Internal Medicine, Karolinska Institute, Danderyd University Hospital, Stockholm, Sweden
| | - Leon Eyrich Jessen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Lyngby, Denmark
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Biomedicum Helsinki, Helsinki, Finland.,Research Programs for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jani K Haukka
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Biomedicum Helsinki, Helsinki, Finland.,Research Programs for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Biomedicum Helsinki, Helsinki, Finland.,Research Programs for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Leif Groop
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden.,Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Mats Eliasson
- Department of Public Health and Clinical Medicine, Sunderby Research Unit, Umeå University, Umeå, Sweden
| | - Björn Eliasson
- Department of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Kerstin Brismar
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Rolf Luft Center for Diabetes Research, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Mahmoud Al-Majdoub
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden
| | - Marja-Riitta Taskinen
- Research Program Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | | | - Peter Spégel
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, 223 62, Lund, Sweden
| | - Tore Julsrud Berg
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Valeriya Lyssenko
- Department of Clinical Science/Diabetes and Endocrinology, Lund University Diabetes Centre, 205 02, Malmö, Sweden. .,Department of Clinical Science, Center for Diabetes Research, University of Bergen, 5032, Bergen, Norway.
| |
Collapse
|
23
|
Tardy AL, Pouteau E, Marquez D, Yilmaz C, Scholey A. Vitamins and Minerals for Energy, Fatigue and Cognition: A Narrative Review of the Biochemical and Clinical Evidence. Nutrients 2020; 12:E228. [PMID: 31963141 PMCID: PMC7019700 DOI: 10.3390/nu12010228] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/17/2022] Open
Abstract
Vitamins and minerals are essential to humans as they play essential roles in a variety of basic metabolic pathways that support fundamental cellular functions. In particular, their involvement in energy-yielding metabolism, DNA synthesis, oxygen transport, and neuronal functions makes them critical for brain and muscular function. These, in turn, translate into effects on cognitive and psychological processes, including mental and physical fatigue. This review is focused on B vitamins (B1, B2, B3, B5, B6, B8, B9 and B12), vitamin C, iron, magnesium and zinc, which have recognized roles in these outcomes. It summarizes the biochemical bases and actions of these micronutrients at both the molecular and cellular levels and connects them with cognitive and psychological symptoms, as well as manifestations of fatigue that may occur when status or supplies of these micronutrients are not adequate.
Collapse
Affiliation(s)
- Anne-Laure Tardy
- Sanofi Consumer Healthcare, Global Medical Nutritionals, 94250 Gentilly, France;
| | - Etienne Pouteau
- Sanofi Consumer Healthcare, Global Medical Nutritionals, 94250 Gentilly, France;
| | | | - Cansu Yilmaz
- Sanofi Consumer Healthcare, 34394 Beşiktaş Istanbul, Turkey;
| | - Andrew Scholey
- Centre for Human Psychopharmacology, Swinburne University, Victoria, VIC 3122, Australia;
| |
Collapse
|
24
|
Gibson GE, Luchsinger JA, Cirio R, Chen H, Franchino-Elder J, Hirsch JA, Bettendorff L, Chen Z, Flowers SA, Gerber LM, Grandville T, Schupf N, Xu H, Stern Y, Habeck C, Jordan B, Fonzetti P. Benfotiamine and Cognitive Decline in Alzheimer's Disease: Results of a Randomized Placebo-Controlled Phase IIa Clinical Trial. J Alzheimers Dis 2020; 78:989-1010. [PMID: 33074237 PMCID: PMC7880246 DOI: 10.3233/jad-200896] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND In preclinical models, benfotiamine efficiently ameliorates the clinical and biological pathologies that define Alzheimer's disease (AD) including impaired cognition, amyloid-β plaques, neurofibrillary tangles, diminished glucose metabolism, oxidative stress, increased advanced glycation end products (AGE), and inflammation. OBJECTIVE To collect preliminary data on feasibility, safety, and efficacy in individuals with amnestic mild cognitive impairment (aMCI) or mild dementia due to AD in a placebo-controlled trial of benfotiamine. METHODS A twelve-month treatment with benfotiamine tested whether clinical decline would be delayed in the benfotiamine group compared to the placebo group. The primary clinical outcome was the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog). Secondary outcomes were the clinical dementia rating (CDR) score and fluorodeoxyglucose (FDG) uptake, measured with brain positron emission tomography (PET). Blood AGE were examined as an exploratory outcome. RESULTS Participants were treated with benfotiamine (34) or placebo (36). Benfotiamine treatment was safe. The increase in ADAS-Cog was 43% lower in the benfotiamine group than in the placebo group, indicating less cognitive decline, and this effect was nearly statistically significant (p = 0.125). Worsening in CDR was 77% lower (p = 0.034) in the benfotiamine group compared to the placebo group, and this effect was stronger in the APOEɛ4 non-carriers. Benfotiamine significantly reduced increases in AGE (p = 0.044), and this effect was stronger in the APOEɛ4 non-carriers. Exploratory analysis derivation of an FDG PET pattern score showed a treatment effect at one year (p = 0.002). CONCLUSION Oral benfotiamine is safe and potentially efficacious in improving cognitive outcomes among persons with MCI and mild AD.
Collapse
Affiliation(s)
- Gary E. Gibson
- Brain and Mind Research Institute, Weil Cornell Medicine, New York, NY, USA
- Burke Neurological Institute, White Plains, NY, USA
| | - José A. Luchsinger
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | - Joseph A. Hirsch
- Burke Neurological Institute, White Plains, NY, USA
- Burke Rehabilitation Hospital, White Plains, NY, USA
- Lenox Hill Hospital, New York, NY, USA
| | - Lucien Bettendorff
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liege, Belgium
| | - Zhengming Chen
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Sarah A. Flowers
- Department of Neuroscience, Georgetown University, Washington, DC, USA
| | - Linda M. Gerber
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | | | - Nicole Schupf
- Mailman School of Public Health, The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Hui Xu
- Burke Neurological Institute, White Plains, NY, USA
| | - Yaakov Stern
- Departments of Neurology, Psychiatry, GH Sergievsky Center, the Taub Institute for the Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Christian Habeck
- Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Barry Jordan
- Rancho Los Amigos National Rehabilitation Center, Downey, CA, USA
- Department of Neurology, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Pasquale Fonzetti
- Einstein College of Medicine, Bronx NY; Westmed Medical Group White Plains NY
| |
Collapse
|
25
|
Vora B, Green EAE, Khuri N, Ballgren F, Sirota M, Giacomini KM. Drug-nutrient interactions: discovering prescription drug inhibitors of the thiamine transporter ThTR-2 (SLC19A3). Am J Clin Nutr 2020; 111:110-121. [PMID: 31764942 PMCID: PMC6944527 DOI: 10.1093/ajcn/nqz255] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/11/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Transporter-mediated drug-nutrient interactions have the potential to cause serious adverse events. However, unlike drug-drug interactions, these drug-nutrient interactions receive little attention during drug development. The clinical importance of drug-nutrient interactions was highlighted when a phase III clinical trial was terminated due to severe adverse events resulting from potent inhibition of thiamine transporter 2 (ThTR-2; SLC19A3). OBJECTIVE In this study, we tested the hypothesis that therapeutic drugs inhibit the intestinal thiamine transporter ThTR-2, which may lead to thiamine deficiency. METHODS For this exploration, we took a multifaceted approach, starting with a high-throughput in vitro primary screen to identify inhibitors, building in silico models to characterize inhibitors, and leveraging real-world data from electronic health records to begin to understand the clinical relevance of these inhibitors. RESULTS Our high-throughput screen of 1360 compounds, including many clinically used drugs, identified 146 potential inhibitors at 200 μM. Inhibition kinetics were determined for 28 drugs with half-maximal inhibitory concentration (IC50) values ranging from 1.03 μM to >1 mM. Several oral drugs, including metformin, were predicted to have intestinal concentrations that may result in ThTR-2-mediated drug-nutrient interactions. Complementary analysis using electronic health records suggested that thiamine laboratory values are reduced in individuals receiving prescription drugs found to significantly inhibit ThTR-2, particularly in vulnerable populations (e.g., individuals with alcoholism). CONCLUSIONS Our comprehensive analysis of prescription drugs suggests that several marketed drugs inhibit ThTR-2, which may contribute to thiamine deficiency, especially in at-risk populations.
Collapse
Affiliation(s)
- Bianca Vora
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Elizabeth A E Green
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Natalia Khuri
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Frida Ballgren
- Department of Pharmaceutical Biosciences, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
26
|
Yu L, Chen Y, Xu Y, He T, Wei Y, He R. D-ribose is elevated in T1DM patients and can be involved in the onset of encephalopathy. Aging (Albany NY) 2019; 11:4943-4969. [PMID: 31307014 PMCID: PMC6682534 DOI: 10.18632/aging.102089] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/04/2019] [Indexed: 12/25/2022]
Abstract
Although many mechanisms have been proposed for diabetic encephalopathy in type 2 diabetes mellitus (T2DM), the risk factors for cognitive impairment in type 1 diabetes mellitus (T1DM) are less clear. Here, we show that streptozotocin (STZ)-induced T1DM rats showed cognitive impairment in both Y maze and Morris water maze assays, accompanied with D-ribose was significantly increased in blood and urine, in addition to D-glucose. Furthermore, advanced glycation end products (AGE), Tau hyperphosphorylation and neuronal death in the hippocampal CA4/DG region were detected in T1DM rats. The expression and activity of transketolase (TKT), an important enzyme in the pentose shunt, were decreased in the brain, indicating that TKT may be involved in D-ribose metabolism in T1DM. Support for these change was demonstrated by the activation of TKT with benfotiamine (BTMP) treatment. Decreased D-ribose levels but not D-glucose levels; markedly reduced AGE accumulation, Tau hyperphosphorylation, and neuronal death; and improved cognitive ability in T1DM rats were shown after BTMP administration. In clinical investigation, T1DM patients had high D-ribose levels in both urine and serum. Our work suggests that D-ribose is involved in the cognitive impairment in T1DM and may provide a potentially novel target for treating diabetic encephalopathy.
Collapse
Affiliation(s)
- Lexiang Yu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yao Chen
- School of Basic Medical Sciences of Southwest Medical University, Luzhou 646000, China
| | - Yong Xu
- Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tao He
- School of Basic Medical Sciences of Southwest Medical University, Luzhou 646000, China
| | - Yan Wei
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing 100101, China
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rongqiao He
- School of Basic Medical Sciences of Southwest Medical University, Luzhou 646000, China
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing 100101, China
- Alzheimer’s Disease Center, Beijing Institute for Brain Disorders, Center for Brain Disorders Research, Capital Medical University, Beijing 100069, China
| |
Collapse
|
27
|
AGE-RAGE stress: a changing landscape in pathology and treatment of Alzheimer's disease. Mol Cell Biochem 2019; 459:95-112. [PMID: 31079281 DOI: 10.1007/s11010-019-03553-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/04/2019] [Indexed: 12/27/2022]
Abstract
Numerous hypotheses including amyloid cascade, cholinergic, and oxidative have been proposed for pathogenesis of Alzheimer's disease (AD). The data suggest that advanced glycation end products (AGEs) and its receptor RAGE (receptor for AGE) are involved in the pathogenesis of AD. AGE-RAGE stress, defined as a balance between stressors (AGE, RAGE) and anti-stressors (sRAGE, AGE degraders) in favor of stressors, has been implicated in pathogenesis of diseases. AGE and its interaction with RAGE-mediated increase in the reactive oxygen species (ROS) damage brain because of its increased vulnerability to ROS. AGE and ROS increase the synthesis of amyloid β (Aβ) leading to deposition of Aβ and phosphorylation of tau, culminating in formation of plaques and neurofibrillary tangles. ROS increase the synthesis of Aβ, high-mobility group box 1(HMGB1), and S100 that interacts with RAGE to produce additional ROS resulting in enhancement of AD pathology. Elevation of ROS precedes the Aβ plaques formation. Because of involvement of AGE and RAGE in AD pathology, the treatment should be targeted at lowering AGE levels through reduction in consumption and formation of AGE, and lowering expression of RAGE, blocking of RAGE ligand binding, increasing levels of soluble RAGE (sRAGE), and use of antioxidants. The above treatment aspect of AD is lacking. In conclusion, AGE-RAGE stress initiates, and Aβ, HMGB1, and S100 enhance the progression of AD. Reduction of levels of AGE and RAGE, elevation of sRAGE, and antioxidants would be beneficial therapeutic modalities in the prevention, regression, and slowing of progression of AD.
Collapse
|
28
|
Gorlova A, Pavlov D, Anthony DC, Ponomarev ED, Sambon M, Proshin A, Shafarevich I, Babaevskaya D, Lesсh KP, Bettendorff L, Strekalova T. Thiamine and benfotiamine counteract ultrasound-induced aggression, normalize AMPA receptor expression and plasticity markers, and reduce oxidative stress in mice. Neuropharmacology 2019; 156:107543. [PMID: 30817932 DOI: 10.1016/j.neuropharm.2019.02.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/08/2019] [Accepted: 02/18/2019] [Indexed: 12/11/2022]
Abstract
The negative societal impacts associated with the increasing prevalence of violence and aggression is increasing, and, with this rise, is the need to understand the molecular and cellular changes that underpin ultrasound-induced aggressive behavior. In mice, stress-induced aggression is known to alter AMPA receptor subunit expression, plasticity markers, and oxidative stress within the brain. Here, we induced aggression in BALB/c mice using chronic ultrasound exposure and examined the impact of the psychoactive anti-oxidant compounds thiamine (vitamin B1), and its derivative benfotiamine, on AMPA receptor subunit expression, established plasticity markers, and oxidative stress. The administration of thiamine or benfotiamine (200 mg/kg/day) in drinking water decreased aggressive behavior following 3-weeks of ultrasound exposure and benfotiamine, reduced floating behavior in the swim test. The vehicle-treated ultrasound-exposed mice exhibited increases in protein carbonyl and total glutathione, altered AMPA receptor subunits expression, and decreased expression of plasticity markers. These ultrasound-induced effects were ameliorated by thiamine and benfotiamine treatment; in particular both antioxidants were able to reverse ultrasound-induced changes in GluA1 and GluA2 subunit expression, and, within the prefrontal cortex, significantly reversed the changes in protein carbonyl and polysialylated form of neural cell adhesion molecule (PSA-NCAM) expression levels. Benfotiamine was usually more efficacious than thiamine. Thus, the thiamine compounds were able to counteract ultrasound-induced aggression, which was accompanied by the normalization of markers that have been showed to be associated with ultrasound-induced aggression. These commonly used, orally-active compounds may have considerable potential for use in the control of aggression within the community. This article is part of the Special Issue entitled 'Current status of the neurobiology of aggression and impulsivity'.
Collapse
Affiliation(s)
- Anna Gorlova
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL, 6229ER, Maastricht, Netherlands; Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, 4000 Liège, Belgium; Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University Trubetskaya Street 8-2, 119991, Moscow, Russia; Department of Biology, Lomonosov Moscow State University, Leninskie Gory1-12, 119991, Moscow, Russia
| | - Dmitrii Pavlov
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL, 6229ER, Maastricht, Netherlands; Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, 4000 Liège, Belgium; Department of Biology, Lomonosov Moscow State University, Leninskie Gory1-12, 119991, Moscow, Russia; Institute of General Pathology and Pathophysiology, Baltiiskaya Str, 8, 125315, Moscow, Russia
| | - Daniel C Anthony
- Department of Pharmacology, Oxford University, Mansfield Road, OX1 3QT, Oxford, United Kingdom
| | - Eugene D Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Margaux Sambon
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, 4000 Liège, Belgium
| | - Andrey Proshin
- Research Institute of Normal Physiology, Baltiiskaya Str, 8, 125315, Moscow, Russia
| | - Igor Shafarevich
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL, 6229ER, Maastricht, Netherlands; Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University Trubetskaya Street 8-2, 119991, Moscow, Russia
| | - Diana Babaevskaya
- Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University Trubetskaya Street 8-2, 119991, Moscow, Russia
| | - Klaus-Peter Lesсh
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL, 6229ER, Maastricht, Netherlands; Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University Trubetskaya Street 8-2, 119991, Moscow, Russia; Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Josef-Schneider-Straße 2, 97080, Wuerzburg, Germany
| | - Lucien Bettendorff
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, 4000 Liège, Belgium.
| | - Tatyana Strekalova
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL, 6229ER, Maastricht, Netherlands; Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University Trubetskaya Street 8-2, 119991, Moscow, Russia; Institute of General Pathology and Pathophysiology, Baltiiskaya Str, 8, 125315, Moscow, Russia.
| |
Collapse
|
29
|
Zhao L, Cheng X, Zhong C. Implications of Successful Symptomatic Treatment in Parkinson's Disease for Therapeutic Strategies of Alzheimer's Disease. ACS Chem Neurosci 2019; 10:922-930. [PMID: 30474958 DOI: 10.1021/acschemneuro.8b00450] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) has been a devastating neurodegenerative disorder and lacks effective treatment to improve the prognosis for patients. Symptomatic treatment for AD mainly includes two categories: Acetylcholinesterase inhibitors and the N-methyl-d-aspartate (NMDA) receptor antagonist (memantine). They cannot significantly improve the quality of life and extend survival time for AD patients. Worse, almost all clinical trials for disease-modifying drugs have failed, and the reduction of brain β-amyloid (Aβ) deposition by multiple approaches, including inhibitors of β- or γ-secretase, vaccines, and antibodies against Aβ deposition, was found to have little effect on AD progression. A new therapeutic strategy for AD is urgently needed. Parkinson's disease also is a neurodegenerative disease having no effective treatment for modifying the disease. Nevertheless, successful symptomatic treatment using the combined therapies of l-DOPA supplement and modulators of l-DOPA metabolism greatly improves the prognosis for PD patients; the average survival time of the patient has been extended from 3-4 years to 10-15 years although dopaminergic neurons are still progressively decreasing. This provides useful implications for AD therapeutic strategies. AD patients manifest global cognitive decline, prominently represented by memory deficit, especially in the early stages of the disease. Further, the degree of decreased cognitive abilities correlates with cholinergic dysfunction and the hypometabolism of glucose, the dominant energy fuel for brain. Thus, the amelioration of brain cholinergic function and brain energy metabolism may be effective treatment to improve cognitive abilities of AD patients. Here, we highlighted the explorations of symptomatic therapeutics through modulating brain cholinergic function and energy metabolism in AD.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Neurology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200111, China
| | - Xiaoqin Cheng
- Department of Neurology, Zhongshan Hospital, The State Key Laboratory of Medical Neurobiology, The Institute of Brain Science, Fudan University, Shanghai 200032, China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, The State Key Laboratory of Medical Neurobiology, The Institute of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
30
|
Yu Q, Liu H, Sang S, Chen L, Zhao Y, Wang Y, Zhong C. Thiamine deficiency contributes to synapse and neural circuit defects. Biol Res 2018; 51:35. [PMID: 30231926 PMCID: PMC6145320 DOI: 10.1186/s40659-018-0184-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/09/2018] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The previous studies have demonstrated the reduction of thiamine diphosphate is specific to Alzheimer's disease (AD) and causal factor of brain glucose hypometabolism, which is considered as a neurodegenerative index of AD and closely correlates with the degree of cognitive impairment. The reduction of thiamine diphosphate may contribute to the dysfunction of synapses and neural circuits, finally leading to cognitive decline. RESULTS To demonstrate this hypothesis, we established abnormalities in the glucose metabolism utilizing thiamine deficiency in vitro and in vivo, and we found dramatically reduced dendrite spine density. We further detected lowered excitatory neurotransmission and impaired hippocampal long-term potentiation, which are induced by TPK RNAi in vitro. Importantly, via treatment with benfotiamine, Aβ induced spines density decrease was considerably ameliorated. CONCLUSIONS These results revealed that thiamine deficiency contributed to synaptic dysfunction which strongly related to AD pathogenesis. Our results provide new insights into pathogenesis of synaptic and neuronal dysfunction in AD.
Collapse
Affiliation(s)
- Qiujian Yu
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
| | - Huimin Liu
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
| | - Shaoming Sang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
- Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032 China
| | - Lulan Chen
- Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032 China
| | - Yingya Zhao
- Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032 China
| | - Yun Wang
- Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032 China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
- Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032 China
| |
Collapse
|
31
|
Prakash S, Joshi H, Patel J. Progressive supranuclear palsy responding to intravenous thiamine: superimposed Wernicke's encephalopathy? BMJ Case Rep 2018; 2018:bcr-2018-226170. [PMID: 30131406 DOI: 10.1136/bcr-2018-226170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Progressive supranuclear palsy (PSP) may be a risk factor for thiamine deficiency. The classic symptoms of PSP (postural instability, supranuclear vertical gaze palsy and dementia) overlap with the clinical triad of Wernicke's encephalopathy (cognitive impairment, gait problems and ocular abnormality). Therefore, superimposed thiamine deficiency in patients with PSP may aggravate the pre-existing symptoms of PSP. Here, we are reporting a 64-year-old woman having supranuclear ocular palsy, gait instability and dementia for the past 2-3 years. The patient fulfilled the diagnostic criteria of PSP. In parallel, she fulfilled the Caine's criteria of Wernicke's encephalopathy. Her serum thiamine level was low. Supplementation of thiamine led to marked improvement in the symptoms which had been present for many years. These symptoms were originally presumed to be due to PSP. This case highlights the needs to identify superimposed thiamine deficiency in patients with PSP.
Collapse
Affiliation(s)
- Sanjay Prakash
- Department of Neurology, Smt BK Shah Medical Institute and Research Centre, Vadodara, Gujarat, India
| | - Hemant Joshi
- Department of Neurology, Smt BK Shah Medical Institute and Research Centre, Vadodara, Gujarat, India
| | - Jay Patel
- Department of Neurology, Smt BK Shah Medical Institute and Research Centre, Vadodara, Gujarat, India
| |
Collapse
|
32
|
Abdul-Muneer PM, Alikunju S, Schuetz H, Szlachetka AM, Ma X, Haorah J. Impairment of Thiamine Transport at the GUT-BBB-AXIS Contributes to Wernicke's Encephalopathy. Mol Neurobiol 2018; 55:5937-5950. [PMID: 29128903 PMCID: PMC9420083 DOI: 10.1007/s12035-017-0811-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/20/2017] [Indexed: 12/25/2022]
Abstract
Wernicke's encephalopathy, a common neurological disease, is caused by thiamine (vitamin B1) deficiency. Neuropathy resulting from thiamine deficiency is a hallmark of Wernicke-Korsakoff syndrome in chronic alcohol users. The underlying mechanisms of this deficiency and progression of neuropathy remain to be understood. To uncover the unknown mechanisms of thiamine deficiency in alcohol abuse, we used chronic alcohol consumption or thiamine deficiency diet ingestion in animal models. Observations from animal models were validated in primary human neuronal culture for neurodegenerative process. We employed radio-labeled bio-distribution of thiamine, qualitative and quantitative analyses of the various biomarkers and neurodegenerative process. In the present studies, we established that disruption of thiamine transport across the intestinal gut blood-brain barrier axis as the cause of thiamine deficiency in the brain for neurodegeneration. We found that reduction in thiamine transport across these interfaces was the cause of reduction in the synthesis of thiamine pyrophosphate (TPP), an active cofactor for pyruvate dehydrogenase E1α (PDHE1α). Our findings revealed that decrease in the levels of PDHE1α cofactors switched on the activation of PD kinase (PDK) in the brain, thereby triggering the neuronal phosphorylation of PDHE1α (p-PDHE1α). Dysfunctional phosphorylated PDHE1α causes the reduction of mitochondrial aerobic respiration that led to neurodegeneration. We concluded that impairment of thiamine transport across the gut-BBB-axis that led to insufficient TPP synthesis was critical to Wernicke-neuropathy, which could be effectively prevented by stabilizing the thiamine transporters.
Collapse
Affiliation(s)
- P M Abdul-Muneer
- Laboratory of Neurovascular Inflammation and Neurodegeneration, Department of Biomedical Engineering, Center for Injury Bio Mechanics, Materials and Medicine, New Jersey Institute of Technology, 111 Lock Street, Newark, NJ, 07102, USA
| | - Saleena Alikunju
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Heather Schuetz
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Adam M Szlachetka
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xiaotang Ma
- Laboratory of Neurovascular Inflammation and Neurodegeneration, Department of Biomedical Engineering, Center for Injury Bio Mechanics, Materials and Medicine, New Jersey Institute of Technology, 111 Lock Street, Newark, NJ, 07102, USA
| | - James Haorah
- Laboratory of Neurovascular Inflammation and Neurodegeneration, Department of Biomedical Engineering, Center for Injury Bio Mechanics, Materials and Medicine, New Jersey Institute of Technology, 111 Lock Street, Newark, NJ, 07102, USA.
| |
Collapse
|
33
|
Sun BL, Li WW, Zhu C, Jin WS, Zeng F, Liu YH, Bu XL, Zhu J, Yao XQ, Wang YJ. Clinical Research on Alzheimer's Disease: Progress and Perspectives. Neurosci Bull 2018; 34:1111-1118. [PMID: 29956105 DOI: 10.1007/s12264-018-0249-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/27/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD), the most common type of dementia, is becoming a major challenge for global health and social care. However, the current understanding of AD pathogenesis is limited, and no early diagnosis and disease-modifying therapy are currently available. During the past year, significant progress has been made in clinical research on the diagnosis, prevention, and treatment of AD. In this review, we summarize the latest achievements, including diagnostic biomarkers, polygenic hazard score, amyloid and tau PET imaging, clinical trials targeting amyloid-beta (Aβ), tau, and neurotransmitters, early intervention, and primary prevention and systemic intervention approaches, and provide novel perspectives for further efforts to understand and cure the disease.
Collapse
Affiliation(s)
- Bin-Lu Sun
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Wei-Wei Li
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Chi Zhu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Wang-Sheng Jin
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Fan Zeng
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yu-Hui Liu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xian-Le Bu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Jie Zhu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiu-Qing Yao
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| |
Collapse
|
34
|
Nie B, Liang S, Jiang X, Duan S, Huang Q, Zhang T, Li P, Liu H, Shan B. An Automatic Method for Generating an Unbiased Intensity Normalizing Factor in Positron Emission Tomography Image Analysis After Stroke. Neurosci Bull 2018; 34:833-841. [PMID: 29876785 DOI: 10.1007/s12264-018-0240-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/12/2018] [Indexed: 10/14/2022] Open
Abstract
Positron emission tomography (PET) imaging of functional metabolism has been widely used to investigate functional recovery and to evaluate therapeutic efficacy after stroke. The voxel intensity of a PET image is the most important indicator of cellular activity, but is affected by other factors such as the basal metabolic ratio of each subject. In order to locate dysfunctional regions accurately, intensity normalization by a scale factor is a prerequisite in the data analysis, for which the global mean value is most widely used. However, this is unsuitable for stroke studies. Alternatively, a specified scale factor calculated from a reference region is also used, comprising neither hyper- nor hypo-metabolic voxels. But there is no such recognized reference region for stroke studies. Therefore, we proposed a totally data-driven automatic method for unbiased scale factor generation. This factor was generated iteratively until the residual deviation of two adjacent scale factors was reduced by < 5%. Moreover, both simulated and real stroke data were used for evaluation, and these suggested that our proposed unbiased scale factor has better sensitivity and accuracy for stroke studies.
Collapse
Affiliation(s)
- Binbin Nie
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Engineering Research Center of Radiographic Techniques and Equipment, Beijing, 100049, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shengxiang Liang
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Engineering Research Center of Radiographic Techniques and Equipment, Beijing, 100049, China.,Physical Science and Technology College, Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaofeng Jiang
- School of Public Health and Family Medicine, Capital Medical University, Beijing, 100069, China
| | - Shaofeng Duan
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Engineering Research Center of Radiographic Techniques and Equipment, Beijing, 100049, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Qi Huang
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Engineering Research Center of Radiographic Techniques and Equipment, Beijing, 100049, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianhao Zhang
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Engineering Research Center of Radiographic Techniques and Equipment, Beijing, 100049, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Panlong Li
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Engineering Research Center of Radiographic Techniques and Equipment, Beijing, 100049, China.,Physical Science and Technology College, Zhengzhou University, Zhengzhou, 450052, China
| | - Hua Liu
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Engineering Research Center of Radiographic Techniques and Equipment, Beijing, 100049, China
| | - Baoci Shan
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China. .,Beijing Engineering Research Center of Radiographic Techniques and Equipment, Beijing, 100049, China. .,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China. .,University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
35
|
Dupont AC, Largeau B, Guilloteau D, Santiago Ribeiro MJ, Arlicot N. The Place of PET to Assess New Therapeutic Effectiveness in Neurodegenerative Diseases. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:7043578. [PMID: 29887768 PMCID: PMC5985069 DOI: 10.1155/2018/7043578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/01/2018] [Indexed: 12/16/2022]
Abstract
In vivo exploration of neurodegenerative diseases by positron emission tomography (PET) imaging has matured over the last 20 years, using dedicated radiopharmaceuticals targeting cellular metabolism, neurotransmission, neuroinflammation, or abnormal protein aggregates (beta-amyloid and intracellular microtubule inclusions containing hyperphosphorylated tau). The ability of PET to characterize biological processes at the cellular and molecular levels enables early detection and identification of molecular mechanisms associated with disease progression, by providing accurate, reliable, and longitudinally reproducible quantitative biomarkers. Thus, PET imaging has become a relevant imaging method for monitoring response to therapy, approved as an outcome measure in bioclinical trials. The aim of this paper is to review and discuss the current inputs of PET in the assessment of therapeutic effectiveness in neurodegenerative diseases connected by common pathophysiological mechanisms, including Parkinson's disease, Huntington's disease, dementia, amyotrophic lateral sclerosis, multiple sclerosis, and also in psychiatric disorders. We also discuss opportunities for PET imaging to drive more personalized neuroprotective and therapeutic strategies, taking into account individual variability, within the growing framework of precision medicine.
Collapse
Affiliation(s)
- Anne-Claire Dupont
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHRU de Tours, Unité de Radiopharmacie, Tours, France
| | | | - Denis Guilloteau
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHRU de Tours, Service de Médecine Nucléaire in vitro, Tours, France
- INSERM CIC 1415, University Hospital, Tours, France
| | - Maria Joao Santiago Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- INSERM CIC 1415, University Hospital, Tours, France
- CHRU de Tours, Service de Médecine Nucléaire in vivo, Tours, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHRU de Tours, Unité de Radiopharmacie, Tours, France
- INSERM CIC 1415, University Hospital, Tours, France
| |
Collapse
|
36
|
Wang C, Fei G, Pan X, Sang S, Wang L, Zhong C, Jin L. High thiamine diphosphate level as a protective factor for Alzheimer's disease. Neurol Res 2018; 40:658-665. [PMID: 29718773 DOI: 10.1080/01616412.2018.1460704] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Objectives Thiamine diphosphate (TDP) is an indispensable coenzyme for three key enzymes in glucose metabolism. Reduced TDP levels in patients with Alzheimer's disease (AD) has been widely demonstrated and is a diagnostic biomarker for the disease. In this study, we further explored the correlation between altered TDP metabolism and AD along with other risk factors. Methods A 1:1 case-control study was employed with 90 AD patients and 90 control subjects with normal-range cognitive abilities as assayed by the Mini Mental Status Evaluation. Age (≤2 years variation), gender, and educational background were strictly matched. Levels of the main thiamine metabolites in whole blood samples, including TDP, thiamine monophosphate, and thiamine, were assayed using high-performance liquid chromatography. Apolipoprotein E genotypes, haemoglobin, and several metabolic factors (fasting glucose, uric acid, triglyceride, and total cholesterol) associated with AD were also measured. Results The odds ratio of TDP level for AD was 0.95 (with TDP level as a continuous variable) or 0.09 (with TDP level as a dichotomized variable with a cut-off value of 99.48 nmol/L). Blood TDP levels were significantly decreased in female AD patients compared to male AD patients. No correlations were identified between TDP levels and several metabolic factors (fasting glucose, uric acid, triglyceride, and total cholesterol). Conclusions TDP is a protective factor for AD and its protective efficacy may be independent of other metabolic factors. The difference of TDP levels between genders may be another possible explanation for the higher prevalence of AD in females.
Collapse
Affiliation(s)
- Changpeng Wang
- a State Key Laboratory of Medical Neurobiology & Collaborative Innovation Center for Brain Science, Department of Neurology , Zhongshan Hospital, Institutes of Brain Science, Fudan University , Shanghai , China
| | - Guoqiang Fei
- a State Key Laboratory of Medical Neurobiology & Collaborative Innovation Center for Brain Science, Department of Neurology , Zhongshan Hospital, Institutes of Brain Science, Fudan University , Shanghai , China
| | - Xiaoli Pan
- a State Key Laboratory of Medical Neurobiology & Collaborative Innovation Center for Brain Science, Department of Neurology , Zhongshan Hospital, Institutes of Brain Science, Fudan University , Shanghai , China
| | - Shaoming Sang
- a State Key Laboratory of Medical Neurobiology & Collaborative Innovation Center for Brain Science, Department of Neurology , Zhongshan Hospital, Institutes of Brain Science, Fudan University , Shanghai , China
| | - Lijun Wang
- a State Key Laboratory of Medical Neurobiology & Collaborative Innovation Center for Brain Science, Department of Neurology , Zhongshan Hospital, Institutes of Brain Science, Fudan University , Shanghai , China
| | - Chunjiu Zhong
- a State Key Laboratory of Medical Neurobiology & Collaborative Innovation Center for Brain Science, Department of Neurology , Zhongshan Hospital, Institutes of Brain Science, Fudan University , Shanghai , China
| | - Lirong Jin
- a State Key Laboratory of Medical Neurobiology & Collaborative Innovation Center for Brain Science, Department of Neurology , Zhongshan Hospital, Institutes of Brain Science, Fudan University , Shanghai , China
| |
Collapse
|
37
|
Etchegoyen M, Nobile MH, Baez F, Posesorski B, González J, Lago N, Milei J, Otero-Losada M. Metabolic Syndrome and Neuroprotection. Front Neurosci 2018; 12:196. [PMID: 29731703 PMCID: PMC5919958 DOI: 10.3389/fnins.2018.00196] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/12/2018] [Indexed: 12/17/2022] Open
Abstract
Introduction: Over the years the prevalence of metabolic syndrome (MetS) has drastically increased in developing countries as a major byproduct of industrialization. Many factors, such as the consumption of high-calorie diets and a sedentary lifestyle, bolster the spread of this disorder. Undoubtedly, the massive and still increasing incidence of MetS places this epidemic as an important public health issue. Hereon we revisit another outlook of MetS beyond its classical association with cardiovascular disease (CVD) and Diabetes Mellitus Type 2 (DM2), for MetS also poses a risk factor for the nervous tissue and threatens neuronal function. First, we revise a few essential concepts of MetS pathophysiology. Second, we explore some neuroprotective approaches in MetS pertaining brain hypoxia. The articles chosen for this review range from the years 1989 until 2017; the selection criteria was based on those providing data and exploratory information on MetS as well as those that studied innovative therapeutic approaches. Pathophysiology: The characteristically impaired metabolic pathways of MetS lead to hyperglycemia, insulin resistance (IR), inflammation, and hypoxia, all closely associated with an overall pro-oxidative status. Oxidative stress is well-known to cause the wreckage of cellular structures and tissue architecture. Alteration of the redox homeostasis and oxidative stress alter the macromolecular array of DNA, lipids, and proteins, in turn disrupting the biochemical pathways necessary for normal cell function. Neuroprotection: Different neuroprotective strategies are discussed involving lifestyle changes, medication aimed to mitigate MetS cardinal symptoms, and treatments targeted toward reducing oxidative stress. It is well-known that the routine practice of physical exercise, aerobic activity in particular, and a complete and well-balanced nutrition are key factors to prevent MetS. Nevertheless, pharmacological control of MetS as a whole and pertaining hypertension, dyslipidemia, and endothelial injury contribute to neuronal health improvement. Conclusion: The development of MetS has risen as a risk factor for neurological disorders. The therapeutic strategies include multidisciplinary approaches directed to address different pathological pathways all in concert.
Collapse
Affiliation(s)
- Melisa Etchegoyen
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Mariana H Nobile
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Francisco Baez
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Barbara Posesorski
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Julian González
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Néstor Lago
- Institute of Cardiovascular Pathophysiology, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - José Milei
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
38
|
Sang S, Pan X, Chen Z, Zeng F, Pan S, Liu H, Jin L, Fei G, Wang C, Ren S, Jiao F, Bao W, Zhou W, Guan Y, Zhang Y, Shi H, Wang Y, Yu X, Wang Y, Zhong C. Thiamine diphosphate reduction strongly correlates with brain glucose hypometabolism in Alzheimer's disease, whereas amyloid deposition does not. ALZHEIMERS RESEARCH & THERAPY 2018; 10:26. [PMID: 29490669 PMCID: PMC5831864 DOI: 10.1186/s13195-018-0354-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 02/05/2018] [Indexed: 01/08/2023]
Abstract
Background The underlying mechanism of brain glucose hypometabolism, an invariant neurodegenerative feature that tightly correlates with cognitive impairment and disease progression of Alzheimer’s disease (AD), remains elusive. Methods Positron emission tomography with 2-[18F]fluoro-2-deoxy-d-glucose (FDG-PET) was used to evaluate brain glucose metabolism, presented as the rate of 2-[18F]fluoro-2-deoxy-d-glucose standardized uptake value ratio (FDG SUVR) in patients with AD or control subjects and in mice with or without thiamine deficiency induced by a thiamine-deprived diet. Brain amyloid-β (Aβ) deposition in patients with clinically diagnosed AD was quantified by performing assays using 11C-Pittsburgh compound B PET. The levels of thiamine metabolites in blood samples of patients with AD and control subjects, as well as in blood and brain samples of mice, were detected by high-performance liquid chromatography with fluorescence detection. Results FDG SUVRs in frontal, temporal, and parietal cortices of patients with AD were closely correlated with the levels of blood thiamine diphosphate (TDP) and cognitive abilities, but not with brain Aβ deposition. Mice on a thiamine-deprived diet manifested a significant decline of FDG SUVRs in multiple brain regions as compared with those in control mice, with magnitudes highly correlating with both brain and blood TDP levels. There were no significant differences in the changes of FDG SUVRs in observed brain regions between amyloid precursor protein/presenilin-1 and wild-type mice following thiamine deficiency. Conclusions We demonstrate, for the first time to our knowledge, in vivo that TDP reduction strongly correlates with brain glucose hypometabolism, whereas amyloid deposition does not. Our study provides new insight into the pathogenesis and therapeutic strategy for AD. Electronic supplementary material The online version of this article (10.1186/s13195-018-0354-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shaoming Sang
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Xiaoli Pan
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Zhichun Chen
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Fan Zeng
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Shumei Pan
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Huimin Liu
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Lirong Jin
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Guoqiang Fei
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Changpeng Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Shuhua Ren
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Fangyang Jiao
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Weiqi Bao
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Weiyan Zhou
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yiqiu Zhang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yanjiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yun Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China.
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Institutes of Brain Science & Collaborative Innovation Center for Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Room 1105, Mingdao Building, 138 Yixueyuan Road, Shanghai, 200032, China.
| |
Collapse
|
39
|
|
40
|
Wang Z, Zhu J. MEMOIR: A Novel System for Neural Lineage Tracing. Neurosci Bull 2017; 33:763-765. [PMID: 28780643 PMCID: PMC5725379 DOI: 10.1007/s12264-017-0161-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/11/2017] [Indexed: 12/20/2022] Open
Affiliation(s)
- Zhifu Wang
- Department of Neurosurgery, Fudan University Huashan Hospital, Shanghai, 200040, China
- National Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and The Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Shanghai, 200120, China
| | - Jianhong Zhu
- Department of Neurosurgery, Fudan University Huashan Hospital, Shanghai, 200040, China.
- National Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and The Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Shanghai, 200120, China.
| |
Collapse
|
41
|
Yu Q, Zhong C. Membrane Aging as the Real Culprit of Alzheimer's Disease: Modification of a Hypothesis. Neurosci Bull 2017; 34:369-381. [PMID: 29177767 DOI: 10.1007/s12264-017-0192-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/05/2017] [Indexed: 01/10/2023] Open
Abstract
Our previous studies proposed that Alzheimer's disease (AD) is a metabolic disorder and hypothesized that abnormal brain glucose metabolism inducing multiple pathophysiological cascades contributes to AD pathogenesis. Aging is one of the great significant risk factors for AD. Membrane aging is first prone to affect the function and structure of the brain by impairing glucose metabolism. We presume that risk factors of AD, including genetic factors (e.g., the apolipoprotein E ε4 allele and genetic mutations) and non-genetic factors (such as fat, diabetes, and cardiac failure) accelerate biomembrane aging and lead to the onset and development of the disease. In this review, we further modify our previous hypothesis to demonstrate "membrane aging" as an initial pathogenic factor that results in functional and structural alterations of membranes and, consequently, glucose hypometabolism and multiple pathophysiological cascades.
Collapse
Affiliation(s)
- Qiujian Yu
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
42
|
Comorbidity Burden of Dementia: A Hospital-Based Retrospective Study from 2003 to 2012 in Seven Cities in China. Neurosci Bull 2017; 33:703-710. [PMID: 29134450 DOI: 10.1007/s12264-017-0193-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 10/30/2017] [Indexed: 12/31/2022] Open
Abstract
Dementia is increasing dramatically and imposes a huge burden on society. To date, there is a lack of data on the health status of patients with dementia in China. In an attempt to investigate the comorbidity burden of dementia patients in China at the national level, we enrolled 2,938 patients with Alzheimer's disease (AD), vascular dementia (VaD), or other types of dementia, who were admitted to tertiary hospitals in seven regions of China from January 2003 to December 2012. The Charlson Comorbidity Index (CCI) was used to evaluate the comorbidity burden of the patients with dementia. Among these patients, 53.4% had AD, 26.3% had VaD, and 20.3% had other types of dementia. The CCI was 3.0 ± 1.9 for all patients, 3.4 ± 1.8 for those with VaD, and 3.0 ± 2.1 for those with AD. The CCI increased with age in all patients, and the length of hospital stay and daily expenses rose with age and CCI. Males had a higher CCI and a longer stay than females. Moreover, patients admitted in the last 5 years of the study had a higher CCI than those admitted in the first 5 years. We found that the comorbidity burden of patients with dementia is heavy. These findings provide a better understanding of the overall health status of dementia patients, and help to increase the awareness of clinicians and policy-makers to improve medical care for patients.
Collapse
|
43
|
Vignisse J, Sambon M, Gorlova A, Pavlov D, Caron N, Malgrange B, Shevtsova E, Svistunov A, Anthony DC, Markova N, Bazhenova N, Coumans B, Lakaye B, Wins P, Strekalova T, Bettendorff L. Thiamine and benfotiamine prevent stress-induced suppression of hippocampal neurogenesis in mice exposed to predation without affecting brain thiamine diphosphate levels. Mol Cell Neurosci 2017; 82:126-136. [PMID: 28506637 DOI: 10.1016/j.mcn.2017.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 12/31/2022] Open
Abstract
Thiamine is essential for normal brain function and its deficiency causes metabolic impairment, specific lesions, oxidative damage and reduced adult hippocampal neurogenesis (AHN). Thiamine precursors with increased bioavailability, especially benfotiamine, exert neuroprotective effects not only for thiamine deficiency (TD), but also in mouse models of neurodegeneration. As it is known that AHN is impaired by stress in rodents, we exposed C57BL6/J mice to predator stress for 5 consecutive nights and studied the proliferation (number of Ki67-positive cells) and survival (number of BrdU-positive cells) of newborn immature neurons in the subgranular zone of the dentate gyrus. In stressed mice, the number of Ki67- and BrdU-positive cells was reduced compared to non-stressed animals. This reduction was prevented when the mice were treated (200mg/kg/day in drinking water for 20days) with thiamine or benfotiamine, that were recently found to prevent stress-induced behavioral changes and glycogen synthase kinase-3β (GSK-3β) upregulation in the CNS. Moreover, we show that thiamine and benfotiamine counteract stress-induced bodyweight loss and suppress stress-induced anxiety-like behavior. Both treatments induced a modest increase in the brain content of free thiamine while the level of thiamine diphosphate (ThDP) remained unchanged, suggesting that the beneficial effects observed are not linked to the role of this coenzyme in energy metabolism. Predator stress increased hippocampal protein carbonylation, an indicator of oxidative stress. This effect was antagonized by both thiamine and benfotiamine. Moreover, using cultured mouse neuroblastoma cells, we show that in particular benfotiamine protects against paraquat-induced oxidative stress. We therefore hypothesize that thiamine compounds may act by boosting anti-oxidant cellular defenses, by a mechanism that still remains to be unveiled. Our study demonstrates, for the first time, that thiamine and benfotiamine prevent stress-induced inhibition of hippocampal neurogenesis and accompanying physiological changes. The present data suggest that thiamine precursors with high bioavailability might be useful as a complementary therapy in several neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | - Anna Gorlova
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Dmitrii Pavlov
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nicolas Caron
- GIGA-Neurosciences, University of Liege, Liege, Belgium
| | | | - Elena Shevtsova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Moscow, Russia
| | - Andrey Svistunov
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Natalyia Markova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Moscow, Russia; Department of Pharmacology, Oxford University, Oxford, UK; Institute of General Pathology and Pathophysiology, Moscow 125 315, Russia; Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Natalyia Bazhenova
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Institute of General Pathology and Pathophysiology, Moscow 125 315, Russia; Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | | | | | - Pierre Wins
- GIGA-Neurosciences, University of Liege, Liege, Belgium
| | - Tatyana Strekalova
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.
| | | |
Collapse
|