1
|
Liu Y, Liu Y, Shi L, Zhang X, Liu K, He S. Selenium ameliorates cognitive impairment through activating BDNF/TrkB pathway. J Trace Elem Med Biol 2025; 88:127599. [PMID: 39837256 DOI: 10.1016/j.jtemb.2025.127599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder that primarily affects older adults. Selenium, an essential micronutrient for humans, plays a crucial role in the body's normal physiological and metabolic processes. A long-term deficiency in selenium intake can lead to various diseases and even contribute to the ageing process. This study aims to explore the ameliorative effect of selenium on cognitive impairment in 3 × Tg-AD mice and to determine if its effects are related to the BDNF/TrkB pathway. METHODS We employed the APP/PS1/tau 3 × Tg-AD mouse model for dietary selenium intervention. Behavioural experiments were conducted to assess learning and memory. Additionally, we measured selenium and GSH-Px levels in whole blood and brain tissue. Neuronal apoptosis in the hippocampus was observed using transmission electron microscopy. The expressions of Aβ, P-tau, BDNF, TrkB, and CREB were measured via RT-qPCR, while the expressions of Aβ, P-tau, BDNF, TrkB, p-CREB, and CREB were quantified using Western blot analysis. RESULTS Our findings indicate that selenium supplementation can improve spatial learning and memory deficiencies in 3 × Tg-AD mice. Selenium supplementation increased selenium and GSH-Px levels in the brain tissue of 3 × Tg-AD mice and significantly enhanced neuronal conditions. Furthermore, the expression levels of proteins related to the BDNF/TrkB pathway significantly increased following selenium supplementation. CONCLUSIONS Our study demonstrates that selenium can ameliorate memory impairment in 3 × Tg-AD mice by activating the BDNF/TrkB pathway.
Collapse
Affiliation(s)
- Yu Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan 750004, China; Department of Medical Administration, Baotou Central Hospital, Baotou, Inner Mongolia 014040, China
| | - Ye Liu
- Infectious Diseases Clinic, The Fourth People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Liping Shi
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan 750004, China
| | - Xue Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan 750004, China
| | - Kunmei Liu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, Yinchuan 750004, China.
| | - Shulan He
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, China.
| |
Collapse
|
2
|
Kara S, Polat S, Akillioglu K, Saker D, Evli Ce AT, Sencar L, Aydın UF, Polat S. Effects of TGF-β1 on Aβ-40 and α- β- γ secretase expression in hippocampus and prefrontal cortex in experimental Alzheimer's disease. Behav Brain Res 2025; 482:115432. [PMID: 39828086 DOI: 10.1016/j.bbr.2025.115432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Alzheimer's disease is a chronic complex neurodegenerative disease characterized with amyloid plaques and loss of neurons. TGF-β1 is important growth factor, plays critical roles in cell metabolism, tissue homeostasis, neuronal development, and synaptic plasticity. In this study, we aimed to examine the effect of TGF-β1 on the regulation of α, β, and γ-secretase enzymes, Aβ-40 accumulation, apoptosis, and neuronal damage in an experimental Scopolamine-induced AD-like model. The subjects were divided into 5 groups such as control, sham, TGF-β1 control, Scopolamin group, TGF-β1 treatment groups.Then all groups were divided into 2 subgroups according to 28th-56th days. Except for Morris water maze (MWM) test, hippocampus and prefrontal cortex tissues were taken for light-electron microscopic, immunohistochemical, and biochemical examinations. It was observed that learning and memory abilities, which decreased in the MWM test of the Scopolamine group, increased in the treatment groups. In addition, α-secretase expression decreased in the Scopolamin group, while it increased in the TGF-β1 treatment group. It was determined that Aβ-40 and caspase-3 immunoreactivity, β and γ-secretase enzyme levels increased in the Scopolamin group and decreased in TGF-β1 treatment group. Cellular degenerations were relatively decreased in TGF-β1 treatment group. It was thought that TGF-β1 might have a therapeutic effect on Alzheimer's disease by increasing memory performance and preventing Aβ-40 accumulation in the AD-like model induced by Scopolamine and also, may be effective preventing neuronal damage by down-regulating caspase-3 expression. When all the findings evaluated together, it was concluded that TGF-β1 could be evaluated as a therapeutic agent in Alzheimer's disease.
Collapse
Affiliation(s)
- Samet Kara
- Department of Histology and Embryology, Faculty of Medicine, University of Çukurova, Adana, Balcali 01330, Turkey.
| | - Sema Polat
- Department of Anatomy, Faculty of Medicine, University of Çukurova, Adana, Balcali 01330, Turkey
| | - Kübra Akillioglu
- Department of Physiology, Faculty of Medicine, University of Çukurova, Adana, Balcali 01330, Turkey
| | - Dilek Saker
- Department of Histology and Embryology, Faculty of Medicine, University of Çukurova, Adana, Balcali 01330, Turkey
| | - Ahmet Turan Evli Ce
- Department of Neurology, Faculty of Medicine, University of Çukurova, Adana, Balcali 01330, Turkey
| | - Leman Sencar
- Department of Histology and Embryology, Faculty of Medicine, University of Çukurova, Adana, Balcali 01330, Turkey
| | - Ummuhan Fulden Aydın
- Department of Biochemistry, Faculty of Medicine, University of Çukurova, Adana, Balcali 01330, Turkey
| | - Sait Polat
- Department of Histology and Embryology, Faculty of Medicine, University of Çukurova, Adana, Balcali 01330, Turkey
| |
Collapse
|
3
|
Elazab ST, Hsu WH. α-Bisabolol alleviates doxorubicin-induced cognitive dysfunction in rats via enhancing the hippocampal BDNF/TrKB signaling and inhibiting neuroinflammation. Front Pharmacol 2025; 16:1549009. [PMID: 40124785 PMCID: PMC11925949 DOI: 10.3389/fphar.2025.1549009] [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: 12/20/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Chemofog is a serious sequela commonly manifested among cancer patients receiving doxorubicin (DOX) chemotherapy. Our goal was to explore the abrogative action of α-Bisabolol (BISA), a phytochemical sesquiterpene, against DOX-induced cognitive deficit. Rats were allocated into 5 groups: Group I: control; Group II received BISA orally (100 mg/kg/day for 4 weeks); Group III received DOX (2 mg/kg/week/i.p.) for 4 weeks; Groups IV and V were administered BISA orally at 50 and 100 mg/kg, respectively plus DOX, i. p. Results: 1) BISA attenuated DOX-induced chemofog as shown in memory-related behavioral tests. 2) BISA restored the hippocampal histological structure and redox homeostasis via diminishing MDA content and upregulating Nrf2 and HO-1 genes. 3) BISA mitigated DOX-induced neuroinflammation through reducing NF-kB, TNF-α, IL-6, IL-1β, and GFAP expressions. 4) BISA repressed the hippocampal apoptosis via downregulating Bax gene and upregulating Bcl-2 gene. 5) BISA enhanced the synaptic plasticity by activating the BDNF/TrKB signaling and increasing the levels of neurotransmitters that enhance memory, i.e., ACh, 5-HT, and DA. BISA at 100 mg/kg/day exerted a better neuroprotection than BISA at 50 mg/kg/day. Thus, BISA may protect cancer patients from cognitive disorders caused by DOX.
Collapse
Affiliation(s)
- Sara T. Elazab
- Department of Pharmacology, Facultyof Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Walter H. Hsu
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
4
|
Mucci F, Arone A, Gurrieri R, Weiss F, Russomanno G, Marazziti D. Third-Generation Antipsychotics: The Quest for the Key to Neurotrophism. Life (Basel) 2025; 15:391. [PMID: 40141736 PMCID: PMC11944073 DOI: 10.3390/life15030391] [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: 12/27/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Antipsychotic drugs (APs) have profoundly changed the treatment landscape for psychiatric disorders, yet their impact on neuroplasticity and neurotrophism remains only partially understood. While second-generation antipsychotics (SGAs) are associated with a better side effect profile than their predecessors, the emergence of third-generation antipsychotics (TGAs)-such as brexpiprazole, cariprazine, lurasidone, iloperidone, lumateperone, pimavanserin, and roluperidone-has prompted renewed interest in their potential neuroprotective and pro-cognitive effects. This review attempts to carefully examine the evidence on the neurotrophic properties of TGAs and their role in modulating brain plasticity by analyzing studies published between 2010 and 2024. Although data remain limited and focused primarily on earlier SGAs, emerging findings suggest that some TGAs may exert positive effects on neuroplastic processes, including the modulation of brain-derived neurotrophic factors (BDNFs) and synaptic architecture. However, robust clinical data on their long-term effects and comparative efficacy are lacking; therefore, further research is necessary to validate their role in preventing neurodegenerative changes and improving cognitive outcomes in patients with psychiatric conditions.
Collapse
Affiliation(s)
- Federico Mucci
- Department of Psychiatry, Lucca Zone, Azienda USL Toscana Nord Ovest, 55100 Lucca, Italy;
| | - Alessandro Arone
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Riccardo Gurrieri
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Francesco Weiss
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Gerardo Russomanno
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| |
Collapse
|
5
|
Yang SA, Park SH, Kim EH, Bae WB, Jhee KH. Amelioration of Astrocytic Dysfunction via AQP4/LRP1 Pathway by Zizania latifolia and Tricin in C6 Cells Exposed to Amyloid β and High-Dose Insulin and in Mice Treated with Scopolamine. J Microbiol Biotechnol 2025; 35:e2412026. [PMID: 40016145 PMCID: PMC11896795 DOI: 10.4014/jmb.2412.12026] [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: 12/15/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 03/01/2025]
Abstract
Zizania latifolia and its bioactive compound tricin have been recognized for their anti-inflammatory, anti-allergic, and anti-aging properties. However, the impact of Z. latifolia extract (ZLE) and tricin on astrocyte dysfunction, particularly related to disruptions in the amyloid β (Aβ) clearance pathway, has not been extensively studied. This research aims to explore the regulatory effects of ZLE and tricin on astroglial dysfunction, utilizing astrocytic differentiated C6 cells (passages 75~85) subjected to Aβ and high-dose insulin, as well as scopolamine-induced mice. Results revealed that ZLE (500 μg/ml) and tricin (1 μg/ml) significantly upregulated the expression of astrocyte proteins GFAP and AQP4, brain-derived neurotrophic factor (BDNF), low-density lipoprotein receptor-related protein 1 (LRP1), and matrix metalloproteinases (MMPs) in C6 cells treated with Aβ and high-dose insulin. Furthermore, oral administration of ZLE (100 and 300 mg/kg) and tricin (0.3 mg/kg) in mice led to an increase in acetylcholine (ACh) levels and upregulation of insulin-degrading enzyme (IDE), LRP1, and MMPs, while reducing the levels of acetylcholinesterase (AChE), Aβ and ApoE4. These findings suggest that ZLE and tricin may ameliorate Aβ and high-dose insulin-induced astrocyte dysfunction in C6 cells and scopolamine-treated mice, potentially through the AQP4/LRP1 pathway.
Collapse
Affiliation(s)
- Seun-Ah Yang
- Department of Food Science and Technology, Keimyung University, Daegu 42601, Republic of Korea
| | - Se-Ho Park
- Department of Food Science and Technology, Keimyung University, Daegu 42601, Republic of Korea
| | - Eun-Hye Kim
- Department of Applied Chemistry, Kumoh National Institute of Technology, Gumi 39177, Republic of Korea
| | - Won-Bin Bae
- Department of Food Science and Technology, Keimyung University, Daegu 42601, Republic of Korea
| | - Kwang-Hwan Jhee
- Department of Applied Chemistry, Kumoh National Institute of Technology, Gumi 39177, Republic of Korea
| |
Collapse
|
6
|
Takamiya A, Radwan A, Christiaens D, Van Cauwenberge M, Vande Casteele T, Laroy M, Vansteelandt K, Sunaert S, Koole M, Van den Stock J, Van Laere K, Bouckaert F, Vandenbulcke M, Emsell L. Gray and white matter differences in the medial temporal lobe in late-life depression: a multimodal PET-MRI investigation. Psychol Med 2025; 55:e10. [PMID: 39901804 PMCID: PMC11968119 DOI: 10.1017/s0033291724003362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 02/05/2025]
Abstract
BACKGROUND Late-life depression (LLD) is characterized by medial temporal lobe (MTL) abnormalities. Although gray matter (GM) and white matter (WM) differences in LLD have been reported, few studies have investigated them concurrently. Moreover, the impact of aetiological factors, such as neurodegenerative and cerebrovascular burden, on tissue differences remains elusive. METHODS This prospective cross-sectional study involved 72 participants, including 33 patients with LLD (mean age 72.2 years, 23 female) and 39 healthy controls (HCs) (mean age 70.6 years, 24 female), who underwent clinical and positron emission tomography (PET)-magnetic resonance imaging (MRI) assessments. High-resolution 3D T1-weighted and T2-weighted FLAIR images were used to assess MTL GM volumes and white matter hyperintensities (WMHs), a proxy for cerebrovascular burden. Diffusion kurtosis imaging metrics derived from multishell diffusion MRI data were analyzed to assess WM microstructure in the following MTL bundles reconstructed using constrained spherical deconvolution tractography: uncinate fasciculus, fornix, and cingulum. Standardized uptake value ratio of 18F-MK-6240 in the MTL was used to assess Alzheimer's disease (AD) type tau accumulation as a proxy for neurodegenerative burden. RESULTS Compared to HCs, patients with LLD showed significantly lower bilateral MTL volumes and WM microstructural differences primarily in the uncinate fasciculi bilaterally and right fornix. In patients with LLD, higher vascular burden, but not tau, was associated with lower MTL volume and more pronounced WM differences. CONCLUSIONS LLD was associated with both GM and WM differences in the MTL. Cerebrovascular disease, rather than AD type tau-mediated neurodegenerative processes, may contribute to brain tissue differences in LLD.
Collapse
Affiliation(s)
- Akihiro Takamiya
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Hills Joint Research Laboratory for Future Preventive Medicine and Wellness, Keio University School of Medicine, TokyoJapan
| | - Ahmed Radwan
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
| | - Daan Christiaens
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Electrical Engineering, EST-PSI, KU Leuven, Leuven, Belgium
| | - Margot Van Cauwenberge
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Thomas Vande Casteele
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Maarten Laroy
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Kristof Vansteelandt
- Department of Neurosciences, Research Group Psychiatry, Neuropsychiatry, Academic Center for ECT and Neuromodulation (AcCENT), University Psychiatric Center KU Leuven, Kortenberg, Belgium
| | - Stefan Sunaert
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Michel Koole
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
| | - Jan Van den Stock
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
- Department of Nuclear Medicine and Molecular Imaging, University Hospitals Leuven, Leuven, Belgium
| | - Filip Bouckaert
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Louise Emsell
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Langhnoja J, Buch L, Pillai P. Neurotrophomodulatory effect of TNF-α through NF-κB in rat cortical astrocytes. Cytotechnology 2025; 77:37. [PMID: 39776978 PMCID: PMC11700960 DOI: 10.1007/s10616-024-00698-z] [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: 04/21/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Tumor necrosis factor alpha (TNF-α) is a well-known pro-inflammatory cytokine originally recognized for its ability to induce apoptosis and cell death. However, recent research has revealed that TNF-α also plays a crucial role as a mediator of cell survival, influencing a wide range of cellular functions. The signaling of TNF-α is mediated through two distinct receptors, TNFR1 and TNFR2, which trigger various intracellular pathways, including NF-κB, JNK, and caspase signaling cascades. Both TNFR1 and TNFR2 are expressed in astrocytes, which are specialized glial cells essential for maintaining the structural and functional integrity of the central nervous system (CNS). Astrocytes support neuronal function by regulating brain homeostasis, maintaining synaptic function, and supplying metabolic substrates. In addition, astrocytes are known to secrete a variety of growth factors and neurotrophins, such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and NT-4/5. These neurotrophins play a critical role in supporting neuronal survival, synaptic plasticity, and myelination within the brain. The present study focuses on the role of TNF-α in modulating neurotrophin expression and secretion in rat cortical astrocytes. We demonstrate that TNF-α induces the upregulation of neurotrophins, particularly NGF and BDNF, in cultured astrocytes. This effect is accompanied by an increase in the expression of their respective receptors (TrkA & TrkB), further suggesting a functional modulation of neurotrophic signaling pathways. Notably, we show that the modulation of neurotrophin expression by TNF-α is mediated via the NF-κB signaling pathway. Additionally, we observed that TNF-α also regulates the secretion levels of NGF and BDNF into the culture media of astrocytes in a dose-dependent manner, indicating that TNF-α can modulate both the production and release of these growth factors. Taken together, our findings highlight a previously underexplored neuroprotective role of TNF-α in astrocytes. Specifically, we propose that TNF-α, through the upregulation of neurotrophins, may contribute to maintaining neuronal health and supporting neuroprotection under disease conditions.
Collapse
Affiliation(s)
- Jaldeep Langhnoja
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| | - Lipi Buch
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| | - Prakash Pillai
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| |
Collapse
|
8
|
Turkez H, Alak G, Ozgeris FB, Cilingir Yeltekin A, Ucar A, Parlak V, Şuţan NA, Atamanalp M. Borax attenuates oxidative stress, inflammation, and apoptosis by modulating Nrf2/ROS balance in acrylamide-induced neurotoxicity in rainbow trout. Drug Chem Toxicol 2025; 48:27-36. [PMID: 38938109 DOI: 10.1080/01480545.2024.2370916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/29/2024]
Abstract
Acrylamide (ACR) can have adverse environmental effects because of its multiple applications. Relevant scientific literatures of the existence of ACR residues in foods following processing steps have raised concern in the biochemistry, chemistry and safety of this vinyl substance. The interest has focused on the hepatotoxicity of ACR in animals and humans and on the ACR content mitigation and its detoxification. Borax (BX), as a naturally occurring antioxidant featured boron compound, was selected in this investigation to assess its possible neuro-protective potential against ACR-induced neurotoxicity. Nrf2 axis signaling pathways and detoxification response to oxidative stress after exposure to ACR in brains of rainbow trout, and the effect of BX application on reducing ACR-induced neurotoxicity were investigated. Rainbow trout were acutely exposed to ACR (12.5 mg/L) alone or simultaneously treated with BX (0.75 mg/L) during 96h. The exposed fish were sampled at 48th and 96th and oxidative stress response endpoints, 8-OHdG, Nrf2, TNF-α, caspase-3, in addition to IL-6 activities and the levels of AChE and BDNF in brain tissues of rainbow trout (Oncorhynchus mykiss) were evaluated. Samples showed decreases in the levels of ACR-mediated biomarkers used to assess neural toxicity (SOD, CAT, GPx, AChE, BDNF, GSH), increased levels of MDA, MPO, DNA damage and apoptosis. ACR disrupted the Nrf2 pathway, and induced neurotoxicity. Inhibited activities' expressions under simultaneous administration experiments, revealed the protective effects of BX against ACR-induced toxicity damage. The obtained data allow the outline of early multi-parameter signaling pathways in rainbow trout.
Collapse
Affiliation(s)
- Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Gonca Alak
- Department of Seaafod Processing, Faculty of Fisheries, Atatürk University, Erzurum, Turkey
| | - Fatma Betul Ozgeris
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Atatürk University, Erzurum, Turkey
| | | | - Arzu Ucar
- Department of Aquaculture, Faculty of Fisheries, Ataturk University, Erzurum, Turkey
| | - Veysel Parlak
- Department of Basic Sciences, Faculty of Fisheries, Ataturk University, Erzurum, Turkey
| | | | - Muhammed Atamanalp
- Department of Aquaculture, Faculty of Fisheries, Ataturk University, Erzurum, Turkey
| |
Collapse
|
9
|
Petkova-Kirova P, Anastassova N, Minchev B, Uzunova D, Grigorova V, Tsvetanova E, Georgieva A, Alexandrova A, Stefanova M, Yancheva D, Kalfin R, Tancheva L. Behavioral and Biochemical Effects of an Arylhydrazone Derivative of 5-Methoxyindole-2-Carboxylic Acid in a Scopolamine-Induced Model of Alzheimer's Type Dementia in Rats. Molecules 2024; 29:5711. [PMID: 39683869 DOI: 10.3390/molecules29235711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) has long proven to be a complex neurodegenerative disorder, with cholinergic dysfunction, oxidative stress, and neuroinflammation being just a few of its pathological features. The complexity of the disease requires a multitargeted treatment covering its many aspects. In the present investigation, an arylhydrazone derivative of 5-methoxyindole-2-carboxylic acid (5MeO), with in vitro strong antioxidant, neuroprotective and monoamine oxidase B-inhibiting effects, was studied in a scopolamine-induced Alzheimer-type dementia in rats. Using behavioral and biochemical methods, we evaluated the effects of 5MeO on learning and memory, and elucidated the mechanisms of these effects. Our experiments demonstrated that 5MeO had a beneficial effect on different types of memory as assessed by the step-through and the Barnes maze tasks. It efficiently restored the decreased by scopolamine brain-derived neurotrophic factor and acetylcholine levels and normalized the increased by scopolamine acetylcholine esterase activity in hippocampus. Most effective 5MeO was in counteracting the induced by scopolamine oxidative stress by decreasing the increased by scopolamine levels of lipid peroxidation and by increasing the reduced by scopolamine catalase activity. Blood biochemical analyses demonstrated a favorable safety profile of 5MeO, prompting further pharmacological studies suggesting 5MeO as a safe and efficient candidate in a multitargeted treatment of AD.
Collapse
Affiliation(s)
- Polina Petkova-Kirova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Neda Anastassova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Building 9, 1113 Sofia, Bulgaria
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria
| | - Borislav Minchev
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Valya Grigorova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Elina Tsvetanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Almira Georgieva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Albena Alexandrova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
- Department of Physiology and Biochemistry, National Sports Academy, Acad. S. Mladenov Str. 21, 1700 Sofia, Bulgaria
| | - Miroslava Stefanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Building 9, 1113 Sofia, Bulgaria
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
- Department of Healthcare, Faculty of Public Health, Healthcare and Sport, South-West University, Ivan Mihailov 66, 2700 Blagoevgrad, Bulgaria
| | - Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| |
Collapse
|
10
|
Kozak Z, Miller CWT. Beyond psychedelics: set and setting in general psychiatric practice. Int Rev Psychiatry 2024; 36:833-840. [PMID: 39980213 DOI: 10.1080/09540261.2024.2419662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/14/2024] [Indexed: 02/22/2025]
Abstract
Psychedelic compounds continue gaining scientific and regulatory traction as potential new treatments for psychiatric disorders. While most psychiatrists will likely not work directly with these compounds, psychedelic research practices provide insights that may improve conventional psychiatric care. Through its emphasis on 'set and setting' (mindset and environment, respectively), psychedelic research highlights the importance of non-pharmacologic factors maximizing therapeutic outcomes. While psychedelics and serotonergic antidepressants are distinctly different in their subjective experience, new findings suggest mechanistic overlap between them. Both have been found to modulate neurotrophins, enhance neuroplasticity, and reopen critical periods of learning, molded by the environmental context in which they are administered. This paper will argue that by integrating insights from psychedelic research (particularly set and setting), depression treatment outcomes in traditional psychiatric settings can improve by optimizing non-pharmacological factors in treatment, including the provision of high-quality psychotherapy.
Collapse
Affiliation(s)
- Zofia Kozak
- Department of Psychiatry, Sheppard Pratt Health System, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
11
|
Jakkamsetti MS, Kolusu AS, Rongala S, Arakareddy BP, Nori LP, Samudrala PK. Saroglitazar, a PPAR α/γ agonist alleviates 3-Nitropropionic acid induced neurotoxicity in rats: Unveiling the underlying mechanisms. Neurotoxicology 2024; 105:131-146. [PMID: 39326639 DOI: 10.1016/j.neuro.2024.09.004] [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: 08/06/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Saroglitazar (SGZ), a peroxisomal proliferated activated receptor α/γ agonist showed neuroprotective effects in various neurodegenerative disorders like Alzheimer's and Parkinson's. However, no studies were performed on Huntington's, so the goal of the current study is to examine the effect of SGZ on Huntington's disease like symptoms induced by 3-Nitropropionic acid. In this protocol, twenty-four rats were divided into four groups, each group consisting of 6 animals. Group 1: The control group received 1 % CMC 10 mg/kg, p.o. for 14 days. Groups 2, 3, and 4 received 3-NP 15 mg/kg, i.p. from Day 1 to Day 7. Groups 3 and 4 received SGZ 5 mg/kg, p.o. and 10 mg/kg, p.o. respectively once daily from day 1 to day 14. Various behavioral tests like OFT, rotarod, hanging wire, narrow beam walk, MWM, and Y-maze were performed. On day-15, the animals were euthanised by cervical dislocation and brain sample were isolated for biochemical and histopathological analysis. Administration of 3-NP showed a significant decrease in motor coordination and cognitive function. Furthermore, 3-NP altered the activity of acetylcholinesterase, anti-oxidant enzymes, Nrf-2, NF-κB, BDNF, CREB levels, and histological features. However, treatment with SGZ showed ameliorative effects in the 3-NP induced neurotoxicity via PPAR α/γ pathway by reducing motor dysfunction, memory impairment, cholinesterase levels, oxidative stress, neuroinflammation. It also enhanced the levels of Nrf-2, BDNF, and CREB expression and improved histological features. In conclusion, treatment with Saroglitazar attenuated Huntington's disease-like symptoms in rats which are induced by 3-NP via activation of PPAR α/γ pathway.
Collapse
Affiliation(s)
- Madhuri Suma Jakkamsetti
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Aravinda Sai Kolusu
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Suma Rongala
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Bhanu Prakash Arakareddy
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Lakshmi Prashanthi Nori
- Department of Pharmaceutics, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India.
| |
Collapse
|
12
|
Mahanta AK, Chaulagain B, Trivedi R, Singh J. Mannose-Functionalized Chitosan-Coated PLGA Nanoparticles for Brain-Targeted Codelivery of CBD and BDNF for the Treatment of Alzheimer's Disease. ACS Chem Neurosci 2024; 15:4021-4032. [PMID: 39377785 PMCID: PMC11870748 DOI: 10.1021/acschemneuro.4c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease causing cognitive and memory decline. AD is characterized by the deposition of amyloid-β and hypophosphorylated forms of tau protein. AD brains are found to be associated with neurodegeneration, oxidative stress, and inflammation. Cannabidiol (CBD) shows neuroprotective, antioxidant, and anti-inflammatory properties and simultaneously reduces amyloid-β production and tau hyperphosphorylation. The brain-derived neurotrophic factor (BDNF) plays a vital role in the development and maintenance of the plasticity of the central nervous system. A decline of BDNF levels in AD patients results in reduced plasticity and neuronal cell death. Current therapeutics against AD are limited to only symptomatic relief, necessitating a therapeutic strategy that reverses cognitive decline. In this scenario, combination therapy of CBD and BDNF could be a fruitful strategy for the treatment of AD. We designed mannose-conjugated chitosan-coated poly(d,l-lactide-co-glycolide (PLGA) (CHTMAN-PLGA) nanoparticles for the codelivery of CBD and BDNF to the brain. Chitosan is modified with mannose to specifically target the glucose transporter-1 (GLUT-1) receptor abundantly present in the blood-brain barrier for selectively delivering therapeutics to the brain. The CBD-encapsulated nanoparticles showed an average hydrodynamic diameter of 306 ± 8.12 nm and a zeta potential of 31.7 ± 1.53 mV. The coated nanoparticles prolonged encapsulated CBD release from the PLGA matrix. The coated nanoparticles exhibited sustained release of CBD for up to 22 days with 91.68 ± 2.91% release of the encapsulated drug. The coated nanoparticles, which had a high positive zeta potential (31.7 ± 1.53 mV), encapsulated the plasmid DNA. The qualitative transfection efficiency was investigated using CHTMAN-PLGA-CBD/pGFP in bEND.3, primary astrocytes, and primary neurons, while the quantitative transfection efficiency of the delivery system was determined using CHTMAN-PLGA-CBD/pBDNF. In vitro, the pBDNF transfection study revealed that the BDNF expression was 4-fold higher for CHTMAN-PLGA-CBD/pBDNF than for naked pBDNF in all of the cell lines. The cytotoxicity and hemocompatibility of the designed nanoparticles were tested in bEND.3 cells and red blood cells, respectively, and the nanoparticles were found to be nontoxic and hemocompatible. Hence, mannose-conjugated chitosan-coated PLGA nanoparticles could be useful as brain-targeting delivery vehicles for the codelivery of CBD and BDNF for possible AD treatment.
Collapse
Affiliation(s)
- Arun Kumar Mahanta
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Bivek Chaulagain
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Riddhi Trivedi
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, United States
| |
Collapse
|
13
|
Wei W, Wu Q, Wang S, Dong C, Shao S, Zhang Z, Zhang X, Zhang X, Kan J, Liu F. Treatment with walnut peptide ameliorates memory impairment in zebrafish and rats: promoting the expression of neurotrophic factors and suppressing oxidative stress. Food Funct 2024; 15:8043-8052. [PMID: 38988249 DOI: 10.1039/d4fo00074a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Walnut peptide, a low molecular weight peptide separated from walnuts by enzymatic hydrolysis, is considered as a potential nutraceutical with a variety of biological activities. In this study, we characterized the walnut peptide prepared by alkaline protease hydrolysis and evaluated its neuroprotective effect in zebrafish and rat models of memory disorders. Series of concentrations of the walnut peptide were orally administered to zebrafish and rats to examine its impact on the behavior and biochemical indicators. The results showed that the oral administration of walnut peptide significantly ameliorated the behavioral performance in zebrafish exposed to bisphenol AF (1 μg mL-1) and rats exposed to alcohol (30% ethanol, 10 mL kg-1). Furthermore, the walnut peptide upregulated the expression of neurotrophic-related molecules in zebrafish, such as the brain-derived neurotrophic factor (BDNF) and the glial cell-derived neurotrophic factor (GDNF). In the rat brain, the walnut peptide increased the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), while dramatically reduced malondialdehyde (MDA) level. Together, these findings elucidated that the walnut peptide might partially offset the declarative memory deficits via regulation of neurotrophic-related molecule expression and promotion of the antioxidant defense ability. Therefore, walnut peptide holds the potential for development into functional foods as a nutritional supplement for the management of certain neurodegenerative disorders.
Collapse
Affiliation(s)
- Wei Wei
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Qiming Wu
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, PR China.
| | - Shuai Wang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Chuanmin Dong
- Institute of Scientific and Technical Information of Heze, Heze, Shandong, 274005, PR China
| | - Shujuan Shao
- Heze Administrative Examination and Approval Service Bureau, Heze, Shandong, 274000, PR China
| | - Zhao Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Xiping Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Xuejun Zhang
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, Shandong, 274108, PR China
| | - Juntao Kan
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, PR China.
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
14
|
Duderstadt Y, Schreiber S, Burtscher J, Schega L, Müller NG, Brigadski T, Braun-Dullaeus RC, Leßmann V, Müller P. Controlled Hypoxia Acutely Prevents Physical Inactivity-Induced Peripheral BDNF Decline. Int J Mol Sci 2024; 25:7536. [PMID: 39062779 PMCID: PMC11276956 DOI: 10.3390/ijms25147536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a crucial mediator of neuronal plasticity. Here, we investigated the effects of controlled normobaric hypoxia (NH) combined with physical inactivity on BDNF blood levels and executive functions. A total of 25 healthy adults (25.8 ± 3.3 years, 15 female) were analyzed in a randomized controlled cross-over study. Each intervention began with a 30 min resting phase under normoxia (NOR), followed by a 90 min continuation of NOR or NH (peripheral oxygen saturation [SpO2] 85-80%). Serum and plasma samples were collected every 15 min. Heart rate and SpO2 were continuously measured. Before and after each exposure, cognitive tests were performed and after 24 h another follow-up blood sample was taken. NH decreased SpO2 (p < 0.001, ηp2 = 0.747) and increased heart rate (p = 0.006, ηp2 = 0.116) significantly. The 30-min resting phase under NOR led to a significant BDNF reduction in serum (p < 0.001, ηp2 = 0.581) and plasma (p < 0.001, ηp2 = 0.362). Continuation of NOR further significantly reduced BDNF after another 45 min (p = 0.018) in serum and after 30 min (p = 0.040) and 90 min (p = 0.005) in plasma. There was no significant BDNF decline under NH. A 24 h follow-up examination showed a significant decline in serum BDNF, both after NH and NOR. Our results show that NH has the potential to counteract physical inactivity-induced BDNF decline. Therefore, our study emphasizes the need for a physically active lifestyle and its positive effects on BDNF. This study also demonstrates the need for a standardized protocol for future studies to determine BDNF in serum and plasma.
Collapse
Affiliation(s)
- Yves Duderstadt
- Division of Cardiology and Angiology, University Hospital Magdeburg, 39120 Magdeburg, Germany; (Y.D.)
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Department of Sports Science, Chair of Health and Physical Activity, Otto-von-Guericke University, 39104 Magdeburg, Germany
| | - Stefanie Schreiber
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), 39120 Magdeburg, Germany
- Division of Neurology, University Hospital Magdeburg, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
- German Center for Mental Health (DZPG), 39120 Magdeburg, Germany
| | - Johannes Burtscher
- Institute of Sports Science, University Innsbruck, 6020 Innsbruck, Austria;
| | - Lutz Schega
- Department of Sports Science, Chair of Health and Physical Activity, Otto-von-Guericke University, 39104 Magdeburg, Germany
| | - Notger G. Müller
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Faculty of Health Sciences Brandenburg, University of Potsdam, 14476 Potsdam, Germany
| | - Tanja Brigadski
- Institute of Physiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Department of Informatics and Microsystems Technology, University of Applied Sciences, 67659 Kaiserslautern, Germany
| | - Rüdiger C. Braun-Dullaeus
- Division of Cardiology and Angiology, University Hospital Magdeburg, 39120 Magdeburg, Germany; (Y.D.)
| | - Volkmar Leßmann
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
- German Center for Mental Health (DZPG), 39120 Magdeburg, Germany
- Institute of Physiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Patrick Müller
- Division of Cardiology and Angiology, University Hospital Magdeburg, 39120 Magdeburg, Germany; (Y.D.)
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), 39120 Magdeburg, Germany
- German Center for Mental Health (DZPG), 39120 Magdeburg, Germany
| |
Collapse
|
15
|
Müller T, Riederer P. The vicious circle between homocysteine, methyl group-donating vitamins and chronic levodopa intake in Parkinson's disease. J Neural Transm (Vienna) 2024; 131:631-638. [PMID: 37329350 DOI: 10.1007/s00702-023-02666-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023]
Abstract
A biomarker for declined methylation capacity is elevation of homocysteine levels. They increase the risk for onset of vascular disease and contribute to progression of chronic neurodegeneration and aging. This narrative review discusses associations between homocysteine, consumption of methyl group-donating vitamins and impact on disease-generating mechanisms in levodopa-treated patients with Parkinson's disease. We conclude to recommend levodopa-treated patients to substitute themselves with methyl group-donating vitamins. This is harmless in terms of application of folic acid, methylcobalamin or hydroxocobalamin. Moreover, we suggest a crucial discussion on the value of the various popular hypotheses on Parkinson's disease-generating mechanisms. Findings from studies with acute levodopa exposure describe oxidative stress generation and impaired methylation capacity, which causes gene dysfunction. Their repeated occurrences contribute to onset of mitochondrial dysfunction, iron enrichment and pathologic protein accumulation in the long term. Current research underestimates these epigenetic, metabolic consequences of chronic levodopa application. Supplementary treatment strategies are recommended to avoid levodopa-related side effects.
Collapse
Affiliation(s)
- Thomas Müller
- Department of Neurology, St. Joseph Hospital Berlin-Weissensee, Gartenstr. 1, 13088, Berlin, Germany.
| | - Peter Riederer
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Margarete-Höppel Platz 1, 97080, Würzburg, Germany
| |
Collapse
|
16
|
Song J. BDNF Signaling in Vascular Dementia and Its Effects on Cerebrovascular Dysfunction, Synaptic Plasticity, and Cholinergic System Abnormality. J Lipid Atheroscler 2024; 13:122-138. [PMID: 38826183 PMCID: PMC11140249 DOI: 10.12997/jla.2024.13.2.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 12/19/2023] [Indexed: 06/04/2024] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia and is characterized by memory impairment, blood-brain barrier disruption, neuronal cell loss, glia activation, impaired synaptic plasticity, and cholinergic system abnormalities. To effectively prevent and treat VaD a good understanding of the mechanisms underlying its neuropathology is needed. Brain-derived neurotrophic factor (BDNF) is an important neurotrophic factor with multiple functions in the systemic circulation and the central nervous system and is known to regulate neuronal cell survival, synaptic formation, glia activation, and cognitive decline. Recent studies indicate that when compared with normal subjects, patients with VaD have low serum BDNF levels and that BDNF deficiency in the serum and cerebrospinal fluid is an important indicator of VaD. Here, we review current knowledge on the role of BDNF signaling in the pathology of VaD, such as cerebrovascular dysfunction, synaptic dysfunction, and cholinergic system impairment.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
17
|
Hakami A, Narasimhan K, Comini G, Thiele J, Werner C, Dowd E, Newland B. Cryogel microcarriers for sustained local delivery of growth factors to the brain. J Control Release 2024; 369:404-419. [PMID: 38508528 DOI: 10.1016/j.jconrel.2024.03.023] [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: 12/15/2023] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
Neurotrophic growth factors such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) have been considered as potential therapeutic candidates for neurodegenerative disorders due to their important role in modulating the growth and survival of neurons. However, clinical translation remains elusive, as their large size hinders translocation across the blood-brain barrier (BBB), and their short half-life in vivo necessitates repeated administrations. Local delivery to the brain offers a potential route to the target site but requires a suitable drug-delivery system capable of releasing these proteins in a controlled and sustained manner. Herein, we develop a cryogel microcarrier delivery system which takes advantage of the heparin-binding properties of GDNF and BDNF, to reversibly bind/release these growth factors via electrostatic interactions. Droplet microfluidics and subzero temperature polymerization was used to create monodisperse cryogels with varying degrees of negative charge and an average diameter of 20 μm. By tailoring the inclusion of 3-sulfopropyl acrylate (SPA) as a negatively charged moiety, the release duration of these two growth factors could be adjusted to range from weeks to half a year. 80% SPA cryogels and 20% SPA cryogels were selected to load GDNF and BDNF respectively, for the subsequent biological studies. Cell culture studies demonstrated that these cryogel microcarriers were cytocompatible with neuronal and microglial cell lines, as well as primary neural cultures. Furthermore, in vivo studies confirmed their biocompatibility after administration into the brain, as well as their ability to deliver, retain and release GDNF and BDNF in the striatum. Overall, this study highlights the potential of using cryogel microcarriers for long-term delivery of neurotrophic growth factors to the brain for neurodegenerative disorder therapeutics.
Collapse
Affiliation(s)
- Abrar Hakami
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK; Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Kaushik Narasimhan
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| | - Giulia Comini
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| | - Julian Thiele
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany; Institute of Chemistry, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK.
| |
Collapse
|
18
|
Jiang D, Liu H, Li T, Zhao S, Yang K, Yao F, Zhou B, Feng H, Wang S, Shen J, Tang J, Zhang YX, Wang Y, Guo C, Tang TS. Agomirs upregulating carboxypeptidase E expression rescue hippocampal neurogenesis and memory deficits in Alzheimer's disease. Transl Neurodegener 2024; 13:24. [PMID: 38671492 PMCID: PMC11046780 DOI: 10.1186/s40035-024-00414-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Adult neurogenesis occurs in the subventricular zone (SVZ) and the subgranular zone of the dentate gyrus in the hippocampus. The neuronal stem cells in these two neurogenic niches respond differently to various physiological and pathological stimuli. Recently, we have found that the decrement of carboxypeptidase E (CPE) with aging impairs the maturation of brain-derived neurotrophic factor (BDNF) and neurogenesis in the SVZ. However, it remains unknown whether these events occur in the hippocampus, and what the role of CPE is in the adult hippocampal neurogenesis in the context of Alzheimer's disease (AD). METHODS In vivo screening was performed to search for miRNA mimics capable of upregulating CPE expression and promoting neurogenesis in both neurogenic niches. Among these, two agomirs were further assessed for their effects on hippocampal neurogenesis in the context of AD. We also explored whether these two agomirs could ameliorate behavioral symptoms and AD pathology in mice, using direct intracerebroventricular injection or by non-invasive intranasal instillation. RESULTS Restoration of CPE expression in the hippocampus improved BDNF maturation and boosted adult hippocampal neurogenesis. By screening the miRNA mimics targeting the 5'UTR region of Cpe gene, we developed two agomirs that were capable of upregulating CPE expression. The two agomirs significantly rescued adult neurogenesis and cognition, showing multiple beneficial effects against the AD-associated pathologies in APP/PS1 mice. Of note, noninvasive approach via intranasal delivery of these agomirs improved the behavioral and neurocognitive functions of APP/PS1 mice. CONCLUSIONS CPE may regulate adult hippocampal neurogenesis via the CPE-BDNF-TrkB signaling pathway. This study supports the prospect of developing miRNA agomirs targeting CPE as biopharmaceuticals to counteract aging- and disease-related neurological decline in human brains.
Collapse
Affiliation(s)
- Dongfang Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hongmei Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Tingting Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Song Zhao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Keyan Yang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Fuwen Yao
- Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bo Zhou
- Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101, China
| | - Haiping Feng
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Sijia Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaqi Shen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jinglan Tang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Present Address: Department of Psychology, UC San Diego, La Jolla, CA, 92093, USA
| | - Yu-Xin Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yun Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Caixia Guo
- Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Tie-Shan Tang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
19
|
Valipour B, Simorgh S, Mirsalehi M, Moradi S, Taghizadeh-Hesary F, Seidkhani E, Akbarnejad Z, Alizadeh R. Improvement of spatial learning and memory deficits by intranasal administration of human olfactory ecto-mesenchymal stem cells in an Alzheimer's disease rat model. Brain Res 2024; 1828:148764. [PMID: 38242524 DOI: 10.1016/j.brainres.2024.148764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024]
Abstract
Mesenchymal stem cells therapy provides a new perspective of therapeutic approaches in the treatment of neurodegenerative diseases. The present study aimed to investigate the effects of intranasally transplanted human "olfactory ecto-mesenchymal stem cells" (OE-MSCs) in Alzheimer's disease (AD) rats. In this study, we isolated OE-MSCs from human olfactory lamina propria and phenotypically characterized them using immunocytochemistry and flow cytometry. The undifferentiated OE-MSCs were transplanted either by intranasal (IN) or intrahippocampal (IH) injection to rat models of AD, which were induced by injecting amyloid-beta (Aβ) intrahippocampally. Behavioral, histological, and molecular assessments were performed after a three-month recovery period. Based on the results, intranasal administration of OE-MSCs significantly reduced Aβ accumulation and neuronal loss, improved learning and memory impairments, and increased levels of BDNF (brain-derived neurotrophic factor) and NMDAR (N-methyl-D-Aspartate receptors) in the AD rat model. These changes were more significant in animals who received OE-MSCs by intranasal injection. The results of this study suggest that OE-MSCs have the potential to enhance cognitive function in AD, possibly mediated by BDNF and the NMDA receptors.
Collapse
Affiliation(s)
- Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran; Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Simorgh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marjan Mirsalehi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Salah Moradi
- Department of Life Science Engineering, Faculty of New Science and Technology, University of Tehran, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Seidkhani
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Akbarnejad
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Rafieh Alizadeh
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Alamin M, Humaira Sultana M, Babarinde IA, Azad AKM, Moni MA, Xu H. Single-cell RNA-seq data analysis reveals functionally relevant biomarkers of early brain development and their regulatory footprints in human embryonic stem cells (hESCs). Brief Bioinform 2024; 25:bbae230. [PMID: 38739758 PMCID: PMC11089419 DOI: 10.1093/bib/bbae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/07/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024] Open
Abstract
The complicated process of neuronal development is initiated early in life, with the genetic mechanisms governing this process yet to be fully elucidated. Single-cell RNA sequencing (scRNA-seq) is a potent instrument for pinpointing biomarkers that exhibit differential expression across various cell types and developmental stages. By employing scRNA-seq on human embryonic stem cells, we aim to identify differentially expressed genes (DEGs) crucial for early-stage neuronal development. Our focus extends beyond simply identifying DEGs. We strive to investigate the functional roles of these genes through enrichment analysis and construct gene regulatory networks to understand their interactions. Ultimately, this comprehensive approach aspires to illuminate the molecular mechanisms and transcriptional dynamics governing early human brain development. By uncovering potential links between these DEGs and intelligence, mental disorders, and neurodevelopmental disorders, we hope to shed light on human neurological health and disease. In this study, we have used scRNA-seq to identify DEGs involved in early-stage neuronal development in hESCs. The scRNA-seq data, collected on days 26 (D26) and 54 (D54), of the in vitro differentiation of hESCs to neurons were analyzed. Our analysis identified 539 DEGs between D26 and D54. Functional enrichment of those DEG biomarkers indicated that the up-regulated DEGs participated in neurogenesis, while the down-regulated DEGs were linked to synapse regulation. The Reactome pathway analysis revealed that down-regulated DEGs were involved in the interactions between proteins located in synapse pathways. We also discovered interactions between DEGs and miRNA, transcriptional factors (TFs) and DEGs, and between TF and miRNA. Our study identified 20 significant transcription factors, shedding light on early brain development genetics. The identified DEGs and gene regulatory networks are valuable resources for future research into human brain development and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Md Alamin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | | | - Isaac Adeyemi Babarinde
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - A K M Azad
- Department of Mathematics and Statistics, College of Science, Imam Muhammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia
| | - Mohammad Ali Moni
- Artificial Intelligence and Cyber Futures Institute, Charles Sturt University, Bathurst, NSW 2795, Australia
| | - Haiming Xu
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
21
|
Mohamed DA, Fouda K, Mabrok HB, El-Shamarka ME, Hamed IM. Sourdough bread as nutritional intervention tool for improvement of cognitive dysfunction in diabetic rats. BMC Nutr 2024; 10:53. [PMID: 38528644 DOI: 10.1186/s40795-024-00861-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND The current research targeted to study the impact of nutritional intervention by two sourdough breads in improvement of cognitive dysfunction in diabetic rats. METHODS Type-2 diabetes was induced in rats by Streptozotocin-Nicotinamide (STZ-NC). Diabetic rats were fed on balanced diet or balanced diet containing 20% of sourdough bread I or II for a month. Lipid profile, oxidative stress, inflammatory markers and cognitive functions were assessed in all rats. Gene expression of brain-derived neurotrophic factor (BDNF) and nuclear respiratory factor 2 (NRF-2) were assessed in hippocampal tissue, while expression of phosphoenol pyruvate carboxy kinase (PEPCK), and glucose transporter 2 (GLUT2) genes were evaluated in hepatic tissue. Chemical composition and fatty acids profile were evaluated in the prepared sourdough bread. RESULTS Sourdough bread II showed higher content of phenolic compounds, fat, fiber and carbohydrates. Fatty acids profile revealed that sourdough bread I was higher in saturated fatty acids (16.08%), while sourdough bread sample II was higher in unsaturated fatty acids (79.33%). Sourdough bread I or II feeding rats' showed significant improvement in hyperglycemia, oxidative stress markers, inflammatory markers, lipid profile, liver and kidney functions in association with improvement in cognitive function. Gene expression of BDNF and NRF2 in hippocampal tissue were increased significantly, while hepatic GLUT2 and PEPCK gene expression were down-regulated in diabetic given sourdough bread I or II. CONCLUSION Sourdough bread II was superior in all the studied parameters. The anti-diabetic effect and protection from cognitive dysfunction of sourdough bread samples may be ascribed to the occurrence of dietary fibers, phenolic compounds, and polyunsaturated fatty acids.
Collapse
Affiliation(s)
- Doha A Mohamed
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo, 12622, Egypt.
| | - Karem Fouda
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Hoda B Mabrok
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Marwa E El-Shamarka
- Toxicology and Narcotics Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Ibrahim M Hamed
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| |
Collapse
|
22
|
Kara M, Sahin S, Rabbani F, Oztas E, Hasbal-Celikok G, Kanımdan E, Kocyigit A, Kanwal A, Wade U, Yakunina A, Di Pierro F, Khan A. An in vitro analysis of an innovative standardized phospholipid carrier-based Melissa officinalis L . extract as a potential neuromodulator for emotional distress and related conditions. Front Mol Biosci 2024; 11:1359177. [PMID: 38545418 PMCID: PMC10965792 DOI: 10.3389/fmolb.2024.1359177] [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: 12/20/2023] [Accepted: 02/14/2024] [Indexed: 11/11/2024] Open
Abstract
Background: Melissa officinalis L. (MO), commonly known as lemon balm, a member of the mint family, is considered a calming herb. In various traditional medicines, it has been utilized to reduce stress and anxiety and promote sleep. A growing body of clinical evidence suggests that MO leaf extract supplementation possesses considerable neuropharmacological properties. However, its possible mechanism of action largely remains unknown. Objective: In the present in vitro studies, we comparatively investigated the central nervous system (CNS)-calming and antioxidative stress properties of an innovative standardized phospholipid carrier-based (Phytosome™) MO extract (Relissa™) vs. an unformulated dry MO extract. Methods: The neuropharmacological effect of the extract was studied in the anti-depressant enzymes γ-aminobutyrate transaminase (GABA-T) and monoamine oxidase A (MAO-A) assays and SH-SY5Y cells brain-derived neurotrophic factor (BDNF) expression assay. The neuroprotective effect of the extract against oxidative stress was assessed in SH-SY5Y cell-based (H2O2-exposed) Total Antioxidant Status (TAS) and Total Reactive Oxygen Species (ROS) assays. The cytotoxic effect of the extract was evaluated using MTT and LDH assays. The extract antioxidant effect was also evaluated in cell-free chemical tests, including TEAC-ABTS, DPPH, Ferric Reducing Antioxidant Power (FRAP), Oxygen Radical Antioxidant Capacity (ORAC), and Hydroxyl Radical Antioxidant Capacity (HORAC) assays. Results: Relissa™ exhibited high GABA-T inhibitory activity, IC50 (mg/mL) = 0.064 vs. unformulated dry MO extract, IC50 (mg/mL) = 0.27. Similar inhibitory effects were also observed for MAO-A. Relissa™ demonstrated an improved neuroprotective antioxidant effect on SH-SY5Y cells against H2O2-induced oxidative stress. Compared to unformulated dry MO extract, Relissa™ exerted high protective effect on H2O2-exposed SH-SY5Y cells, leading to higher cells BDNF expression levels. Moreover, cell-free chemical tests, including TEAC-ABTS, DPPH radical scavenging, FRAP, ORAC, and HORAC assays, validated the improved antioxidant effect of Relissa™ vs. unformulated dry MO extract. Conclusion: The results of the present study support the neuromodulating and neuroprotective properties of Relissa™, and its supplementation may help in the amelioration of emotional distress and related conditions.
Collapse
Affiliation(s)
- Mehtap Kara
- Department of Pharmaceutical Toxicology, Istanbul University Faculty of Pharmacy, Istanbul, Türkiye
| | - Sumeyye Sahin
- Department of Food Engineering, Ordu University, Ordu, Türkiye
| | - Fazle Rabbani
- Department of Psychiatry, Lady Reading Hospital, Peshawar, Pakistan
| | - Ezgi Oztas
- Department of Pharmaceutical Toxicology, Istanbul University Faculty of Pharmacy, Istanbul, Türkiye
| | - Gozde Hasbal-Celikok
- Department of Biochemistry, Istanbul University Faculty of Pharmacy, Istanbul, Türkiye
| | - Ebru Kanımdan
- Department of Medical Biochemistry, Bezmialem Vakif University, Istanbul, Türkiye
| | - Abdurrahim Kocyigit
- Department of Medical Biochemistry, Bezmialem Vakif University, Istanbul, Türkiye
| | - Ayesha Kanwal
- Department of Psychiatry, Lady Reading Hospital, Peshawar, Pakistan
| | - Ursula Wade
- Department of Basic and Clinical Neuroscience, Kings College London, London, United Kingdom
| | - Anastasia Yakunina
- Department of Basic and Clinical Neuroscience, Kings College London, London, United Kingdom
| | - Francesco Di Pierro
- Scientific and Research Department, Velleja Research, Milan, Italy
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Amjad Khan
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, Liaquat University of Medical and Health Sciences, Jamshoro, Pakistan
| |
Collapse
|
23
|
Singh M, Krishnamoorthy VR, Kim S, Khurana S, LaPorte HM. Brain-derived neuerotrophic factor and related mechanisms that mediate and influence progesterone-induced neuroprotection. Front Endocrinol (Lausanne) 2024; 15:1286066. [PMID: 38469139 PMCID: PMC10925611 DOI: 10.3389/fendo.2024.1286066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024] Open
Abstract
Historically, progesterone has been studied significantly within the context of reproductive biology. However, there is now an abundance of evidence for its role in regions of the central nervous system (CNS) associated with such non-reproductive functions that include cognition and affect. Here, we describe mechanisms of progesterone action that support its brain-protective effects, and focus particularly on the role of neurotrophins (such as brain-derived neurotrophic factor, BDNF), the receptors that are critical for their regulation, and the role of certain microRNA in influencing the brain-protective effects of progesterone. In addition, we describe evidence to support the particular importance of glia in mediating the neuroprotective effects of progesterone. Through this review of these mechanisms and our own prior published work, we offer insight into why the effects of a progestin on brain protection may be dependent on the type of progestin (e.g., progesterone versus the synthetic, medroxyprogesterone acetate) used, and age, and as such, we offer insight into the future clinical implication of progesterone treatment for such disorders that include Alzheimer's disease, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | | | | | | | | |
Collapse
|
24
|
Babington S, Tilbrook AJ, Maloney SK, Fernandes JN, Crowley TM, Ding L, Fox AH, Zhang S, Kho EA, Cozzolino D, Mahony TJ, Blache D. Finding biomarkers of experience in animals. J Anim Sci Biotechnol 2024; 15:28. [PMID: 38374201 PMCID: PMC10877933 DOI: 10.1186/s40104-023-00989-z] [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: 09/07/2023] [Accepted: 12/28/2023] [Indexed: 02/21/2024] Open
Abstract
At a time when there is a growing public interest in animal welfare, it is critical to have objective means to assess the way that an animal experiences a situation. Objectivity is critical to ensure appropriate animal welfare outcomes. Existing behavioural, physiological, and neurobiological indicators that are used to assess animal welfare can verify the absence of extremely negative outcomes. But welfare is more than an absence of negative outcomes and an appropriate indicator should reflect the full spectrum of experience of an animal, from negative to positive. In this review, we draw from the knowledge of human biomedical science to propose a list of candidate biological markers (biomarkers) that should reflect the experiential state of non-human animals. The proposed biomarkers can be classified on their main function as endocrine, oxidative stress, non-coding molecular, and thermobiological markers. We also discuss practical challenges that must be addressed before any of these biomarkers can become useful to assess the experience of an animal in real-life.
Collapse
Affiliation(s)
- Sarah Babington
- School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Alan J Tilbrook
- Centre for Animal Science, The Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
- School of Veterinary Science, The University of Queensland, Gatton, QLD, 4343, Australia
| | - Shane K Maloney
- School of Human Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Jill N Fernandes
- School of Veterinary Science, The University of Queensland, Gatton, QLD, 4343, Australia
| | - Tamsyn M Crowley
- School of Medicine, Deakin University, Geelong, VIC, 3217, Australia
- Poultry Hub Australia, University of New England, Armidale, NSW, 2350, Australia
| | - Luoyang Ding
- School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Archa H Fox
- School of Human Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Song Zhang
- School of Human Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Elise A Kho
- Centre for Animal Science, The Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Daniel Cozzolino
- Centre for Nutrition and Food Sciences, The Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Timothy J Mahony
- Centre for Animal Science, The Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Dominique Blache
- School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia.
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
25
|
Mattson MP, Leak RK. The hormesis principle of neuroplasticity and neuroprotection. Cell Metab 2024; 36:315-337. [PMID: 38211591 DOI: 10.1016/j.cmet.2023.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/06/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024]
Abstract
Animals live in habitats fraught with a range of environmental challenges to their bodies and brains. Accordingly, cells and organ systems have evolved stress-responsive signaling pathways that enable them to not only withstand environmental challenges but also to prepare for future challenges and function more efficiently. These phylogenetically conserved processes are the foundation of the hormesis principle, in which single or repeated exposures to low levels of environmental challenges improve cellular and organismal fitness and raise the probability of survival. Hormetic principles have been most intensively studied in physical exercise but apply to numerous other challenges known to improve human health (e.g., intermittent fasting, cognitive stimulation, and dietary phytochemicals). Here we review the physiological mechanisms underlying hormesis-based neuroplasticity and neuroprotection. Approaching natural resilience from the lens of hormesis may reveal novel methods for optimizing brain function and lowering the burden of neurological disorders.
Collapse
Affiliation(s)
- Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| |
Collapse
|
26
|
Dadkhah M, Baziar M, Rezaei N. The regulatory role of BDNF in neuroimmune axis function and neuroinflammation induced by chronic stress: A new therapeutic strategies for neurodegenerative disorders. Cytokine 2024; 174:156477. [PMID: 38147741 DOI: 10.1016/j.cyto.2023.156477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/14/2023] [Accepted: 12/10/2023] [Indexed: 12/28/2023]
Abstract
Neurodegenerative disorders account for a high proportion of neurological diseases that significantly threaten public health worldwide. Various factors are involved in the pathophysiology of such diseases which can lead to neurodegeneration and neural damage. Furthermore, neuroinflammation is a well-known factor in predisposing factors of neurological and especially neurodegenerative disorders which can be strongly suppressed by "anti-inflammatory" actions of brain-derived neurotrophic factor (BDNF). Stress has has also been identified as a risk factor in developing neurodegenerative disorders potentially leading to increased neuroinflammation in the brain and progressive loss in neuronal structures and impaired functions in the CNS. Recently, more studies have increasingly been focused on the role of neuroimmune system in regulating the neurobiology of stress. Emerging evidence indicate that exposure to chronic stress might alter the susceptibility to neurodegeneration via influencing the microglia function. Microglia is considered as the first responding group of cells in suppressing neuroinflammation, leading to an increased inflammatory cytokine signaling that promote the synaptic plasticity deficiencies, impairment in neurogenesis, and development of neurodegenerative disorders. In this review we discuss how exposure to chronic stress might alter the neuroimmune response potentially leading to progress of neurodegenerative disorders. We also emphasize on the role of BDNF in regulating the neuroimmune axis function and microglia modulation in neurodegenerative disorders.
Collapse
Affiliation(s)
- Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Milad Baziar
- Student Research Committee, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran 1419733151, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education Research Network (USERN), Tehran, Iran
| |
Collapse
|
27
|
Chowdhury MA, Collins JM, Gell DA, Perry S, Breadmore MC, Shigdar S, King AE. Isolation and Identification of the High-Affinity DNA Aptamer Target to the Brain-Derived Neurotrophic Factor (BDNF). ACS Chem Neurosci 2024; 15:346-356. [PMID: 38149631 DOI: 10.1021/acschemneuro.3c00661] [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] [Indexed: 12/28/2023] Open
Abstract
Aptamers are functional oligonucleotide ligands used for the molecular recognition of various targets. The natural characteristics of aptamers make them an excellent alternative to antibodies in diagnostics, therapeutics, and biosensing. DNA aptamers are mainly single-stranded oligonucleotides (ssDNA) that possess a definite binding to targets. However, the application of aptamers to the fields of brain health and neurodegenerative diseases has been limited to date. Herein, a DNA aptamer against the brain-derived neurotrophic factor (BDNF) protein was obtained by in vitro selection. BDNF is a potential biomarker of brain health and neurodegenerative diseases and has functions in the synaptic plasticity and survival of neurons. We identified eight aptamers that have binding affinity for BDNF from a 50-nucleotide library. Among these aptamers, NV_B12 showed the highest sensitivity and selectivity for detecting BDNF. In an aptamer-linked immobilized sorbent assay (ALISA), the NV_B12 aptamer strongly bound to BDNF protein, in a dose-dependent manner. The dissociation constant (Kd) for NV_B12 was 0.5 nM (95% CI: 0.4-0.6 nM). These findings suggest that BDNF-specific aptamers could be used as an alternative to antibodies in diagnostic and detection assays for BDNF.
Collapse
Affiliation(s)
- Md Anisuzzaman Chowdhury
- Wicking Dementia Research and Education Centre, University of Tasmania, 17 Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Jessica M Collins
- Wicking Dementia Research and Education Centre, University of Tasmania, 17 Liverpool Street, Hobart, Tasmania 7000, Australia
| | - David A Gell
- Menzies Research Institute, School of Medicine, University of Tasmania, 17 Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, University of Tasmania, 17 Liverpool Street, Hobart, Tasmania 7000, Australia
| | - Michael C Breadmore
- Australian Centre for Research on Separation Science (ACROSS), School of Chemistry, University of Tasmania, Sandy Bay, Hobart, Tasmania 7001, Australia
| | - Sarah Shigdar
- School of Medicine, Faculty of Health, Deakin University, Geelong, Victoria 3220, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, 17 Liverpool Street, Hobart, Tasmania 7000, Australia
| |
Collapse
|
28
|
Pettrey C, Kerr PL, Dickey TO. Physical Exercise as an Intervention for Depression: Evidence for Efficacy and Mu-Opioid Receptors as a Mechanism of Action. ADVANCES IN NEUROBIOLOGY 2024; 35:221-239. [PMID: 38874725 DOI: 10.1007/978-3-031-45493-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Physical exercise is often cited as an important part of an intervention for depression, and there is empirical evidence to support this. However, the mechanism of action through which any potential antidepressant effects are produced is not widely understood. Recent evidence points toward the involvement of endogenous opioids, and especially the mu-opioid system, as a partial mediator of these effects. In this chapter, we discuss the current level of empirical support for physical exercise as either an adjunctive or standalone intervention for depression. We then review the extant evidence for involvement of endogenous opioids in the proposed antidepressant effects of exercise, with a focus specifically on evidence for mu-opioid system involvement.
Collapse
Affiliation(s)
| | - Patrick L Kerr
- Behavioral Medicine & Psychiatry, WVU School of Medicine, Charleston, WV, USA
| | - T O Dickey
- West Virginia University School of Medicine-Charleston, Charleston, WV, USA
| |
Collapse
|
29
|
Lalonde R, Hernandez M, Strazielle C. BDNF and Cerebellar Ataxia. Curr Drug Res Rev 2024; 16:300-307. [PMID: 37609676 DOI: 10.2174/2589977515666230811093021] [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: 04/14/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 08/24/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has been proposed as a treatment for neurodegeneration, including diseases of the cerebellum, where BDNF levels or those of its main receptor, TrkB, are often diminished relative to controls, thereby serving as replacement therapy. Experimental evidence indicates that BDNF signaling countered cerebellar degeneration, sensorimotor deficits, or both, in transgenic ATXN1 mice mutated for ataxin-1, Cacna1a knock-in mice mutated for ataxin-6, mice injected with lentivectors encoding RNA sequences against human FXN into the cerebellar cortex, Kcnj6Wv (Weaver) mutant mice with granule cell degeneration, and rats with olivocerebellar transaction, similar to a BDNF-overexpressing transgenic line interbred with Cacng2stg mutant mice. In this regard, this study discusses whether BDNF is effective in cerebellar pathologies where BDNF levels are normal and whether it is effective in cases with combined cerebellar and basal ganglia damage.
Collapse
Affiliation(s)
- Robert Lalonde
- Université de Lorraine, Laboratoire Stress, Immunité, Pathogènes EA 7300, Campus Santé, 9 avenue de la Forêt de Haye, 54500 Vandoeuvre-les-Nancy, France
| | - Magali Hernandez
- Université de Lorraine, Laboratoire Stress, Immunité, Pathogènes EA 7300, Campus Santé, 9 avenue de la Forêt de Haye, 54500 Vandoeuvre-les-Nancy, France
- CHRU Nancy, allée du Morvan, 54500 Vandoeuvre-les-Nancy, France
| | - Catherine Strazielle
- Université de Lorraine, Laboratoire Stress, Immunité, Pathogènes EA 7300, Campus Santé, 9 avenue de la Forêt de Haye, 54500 Vandoeuvre-les-Nancy, France
- CHRU Nancy, allée du Morvan, 54500 Vandoeuvre-les-Nancy, France
| |
Collapse
|
30
|
Wang Y, Liang J, Xu B, Yang J, Wu Z, Cheng L. TrkB/BDNF signaling pathway and its small molecular agonists in CNS injury. Life Sci 2024; 336:122282. [PMID: 38008209 DOI: 10.1016/j.lfs.2023.122282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 10/19/2023] [Accepted: 11/18/2023] [Indexed: 11/28/2023]
Abstract
As one of the most prevalent neurotrophic factors in the central nervous system (CNS), brain-derived neurotrophic factor (BDNF) plays a significant role in CNS injury by binding to its specific receptor Tropomyosin-related kinase receptor B (TrkB). The BDNF/TrkB signaling pathway is crucial for neuronal survival, structural changes, and plasticity. BDNF acts as an axonal growth and extension factor, a pro-survival factor, and a synaptic modulator in the CNS. BDNF also plays an important role in the maintenance and plasticity of neuronal circuits. Several studies have demonstrated the importance of BDNF in the treatment and recovery of neurodegenerative and neurotraumatic disorders. By undertaking in-depth study on the mechanism of BDNF/TrkB function, important novel therapeutic strategies for treating neuropsychiatric disorders have been discovered. In this review, we discuss the expression patterns and mechanisms of the TrkB/BDNF signaling pathway in CNS damage and introduce several intriguing small molecule TrkB receptor agonists produced over the previous several decades.
Collapse
Affiliation(s)
- Yujin Wang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jing Liang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; School of Stomatology, Tongji University, Shanghai 200072, China
| | - Boyu Xu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jin Yang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Zhourui Wu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China.
| |
Collapse
|
31
|
Deng C, Chen H. Brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling in spinal muscular atrophy and amyotrophic lateral sclerosis. Neurobiol Dis 2024; 190:106377. [PMID: 38092270 DOI: 10.1016/j.nbd.2023.106377] [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: 09/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023] Open
Abstract
Tropomyosin receptor kinase B (TrkB) and its primary ligand brain-derived neurotrophic factor (BDNF) are expressed in the neuromuscular system, where they affect neuronal survival, differentiation, and functions. Changes in BDNF levels and full-length TrkB (TrkB-FL) signaling have been revealed in spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS), two common forms of motor neuron diseases that are characterized by defective neuromuscular junctions in early disease stages and subsequently progressive muscle weakness. This review summarizes the current understanding of BDNF/TrkB-FL-related research in SMA and ALS, with an emphasis on their alterations in the neuromuscular system and possible BDNF/TrkB-FL-targeting therapeutic strategies. The limitations of current studies and future directions are also discussed, giving the hope of discovering novel and effective treatments.
Collapse
Affiliation(s)
- Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
32
|
Ricci V, de Berardis D, Martinotti G, Maina G. Neurotrophic Factors in Cannabis-induced Psychosis: An Update. Curr Top Med Chem 2024; 24:1757-1772. [PMID: 37644743 DOI: 10.2174/1568026623666230829152150] [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: 06/12/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Cannabis is the most widely used illicit substance. Numerous scientific evidence confirm the strong association between cannabis and psychosis. Exposure to cannabis can induce the development of psychosis and schizophrenia in vulnerable individuals. However, the neurobiological processes underlying this relationship are unknown. Neurotrophins are a class of proteins that serve as survival factors for central nervous system (CNS) neurons. In particular, Nerve Growth Factor (NGF) plays an important role in the survival and function of cholinergic neurons while Brain Derived Neurotrophic Factor (BDNF) is involved in synaptic plasticity and the maintenance of midbrain dopaminergic and cholinergic neurons. Glial Cell Derived Neurotrophic Factor (GDNF) promotes the survival of midbrain dopaminergic neurons and Neuregulin 1 (NrG- 1) contributes to glutamatergic signals regulating the N-methyl-D-aspartate (NMDA). They have a remarkable influence on the neurons involved in the Δ-9-THC (tethra-hydro-cannabinol) action, such as dopaminergic and glutamatergic neurons, and can play dual roles: first, in neuronal survival and death, and, second, in activity-dependent plasticity. METHODS In this brief update, reviewing in a narrative way the relevant literature, we will focus on the effects of cannabis on this class of proteins, which may be implicated, at least in part, in the mechanism of the psychostimulant-induced neurotoxicity and psychosis. CONCLUSION Since altered levels of neurotrophins may participate in the pathogenesis of psychotic disorders which are common in drug users, one possible hypothesis is that repeated cannabis exposure can cause psychosis by interfering with neurotrophins synthesis and utilization by CNS neurons.
Collapse
Affiliation(s)
- Valerio Ricci
- Psychiatric Service for Diagnosis and Treatment, San Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Turin, Italy
| | - Domenico de Berardis
- NHS, Department of Mental Health, Psychiatric Service for Diagnosis and Treatment, Hospital "G. Mazzini", ASL 4, 64100, Teramo, Italy
| | - Giovanni Martinotti
- Department of Neurosciences, Imaging and Clinical Sciences, University of Chieti-Pescara, 66100, Chieti, Italy
| | - Giuseppe Maina
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Italy
| |
Collapse
|
33
|
Aurelian S, Ciobanu A, Cărare R, Stoica SI, Anghelescu A, Ciobanu V, Onose G, Munteanu C, Popescu C, Andone I, Spînu A, Firan C, Cazacu IS, Trandafir AI, Băilă M, Postoiu RL, Zamfirescu A. Topical Cellular/Tissue and Molecular Aspects Regarding Nonpharmacological Interventions in Alzheimer's Disease-A Systematic Review. Int J Mol Sci 2023; 24:16533. [PMID: 38003723 PMCID: PMC10671501 DOI: 10.3390/ijms242216533] [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: 09/22/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
One of the most complex and challenging developments at the beginning of the third millennium is the alarming increase in demographic aging, mainly-but not exclusively-affecting developed countries. This reality results in one of the harsh medical, social, and economic consequences: the continuously increasing number of people with dementia, including Alzheimer's disease (AD), which accounts for up to 80% of all such types of pathology. Its large and progressive disabling potential, which eventually leads to death, therefore represents an important public health matter, especially because there is no known cure for this disease. Consequently, periodic reappraisals of different therapeutic possibilities are necessary. For this purpose, we conducted this systematic literature review investigating nonpharmacological interventions for AD, including their currently known cellular and molecular action bases. This endeavor was based on the PRISMA method, by which we selected 116 eligible articles published during the last year. Because of the unfortunate lack of effective treatments for AD, it is necessary to enhance efforts toward identifying and improving various therapeutic and rehabilitative approaches, as well as related prophylactic measures.
Collapse
Affiliation(s)
- Sorina Aurelian
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Gerontology and Geriatrics Clinic Division, St. Luca Hospital for Chronic Illnesses, 041915 Bucharest, Romania
| | - Adela Ciobanu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| | - Roxana Cărare
- Faculty of Medicine, University of Southampton, Southampton SO16 7NS, UK;
| | - Simona-Isabelle Stoica
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Aurelian Anghelescu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Vlad Ciobanu
- Computer Science Department, Politehnica University of Bucharest, 060042 Bucharest, Romania;
| | - Gelu Onose
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Constantin Munteanu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Cristina Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Ioana Andone
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Aura Spînu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Carmen Firan
- NeuroRehabilitation Compartment, The Physical and Rehabilitation Medicine & Balneology Clinic Division, Teaching Emergency Hospital of the Ilfov County, 022104 Bucharest, Romania;
| | - Ioana Simona Cazacu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Andreea-Iulia Trandafir
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Mihai Băilă
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Ruxandra-Luciana Postoiu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Andreea Zamfirescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Gerontology and Geriatrics Clinic Division, St. Luca Hospital for Chronic Illnesses, 041915 Bucharest, Romania
| |
Collapse
|
34
|
Salari Z, Ashabi G, Fartoosi A, Fartoosi A, Shariatpanahi M, Aghsami M, Montazeri H, Kheradmand A. Sericin alleviates motor dysfunction by modulating inflammation and TrkB/BDNF signaling pathway in the rotenone-induced Parkinson's disease model. BMC Pharmacol Toxicol 2023; 24:60. [PMID: 37936189 PMCID: PMC10631121 DOI: 10.1186/s40360-023-00703-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the degeneration of nigrostriatal dopaminergic neurons and movement impairment. Based on theories, neuroinflammatory processes may be vital in the etiology of PD and other neurodegenerative diseases. Reports show that rotenone has neurotoxic, inflammatory, and motor impairment effects in PD. Sericin is a natural polymer with effective properties, such as neuroprotective and anti-inflammatory. Therefore, this study aimed to examine the effects of sericin administration on motor dysfunction by modulating inflammation and tyrosine kinase B/brain-derived neurotrophic factor (TrkB/BDNF) pathway in the rotenone-induced PD model. METHODS Wistar male rats (3-months-old) were treated with rotenone (2 mg/kg every 48 h for 30 days) to induce a rotenone-induced PD model. Also, sericin was administered orally at dose of 200 mg/kg every 48 h for 30 days. Rotarod and bar tests were performed for motor dysfunction. The protein levels of BDNF, c-fos, TrkB, tumor necrosis factor- α (TNF-α), interleukin-6 (IL-6) and catalase activity were evaluated in the striatum area. RESULTS Results showed that sericin increased latent time in the rotarod test and decreased the time staying on the pole in the bar test compared to the PD group (P < 0.001 for both tests). Moreover, sericin treatments decreased TNF-α (P < 0.001) and IL-6 (P < 0.001) concentration levels and enhanced the levels of BDNF (P < 0.001), c-fos (P < 0.001), TrkB (P < 0.001) proteins and catalase activity (P < 0.05) in the striatum area compared to the PD group. CONCLUSION These results support a protective benefit of sericin therapy in a rotenone-induced PD paradigm by reducing motor impairment, inflammatory response, and disruption of the TrkB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Zahra Salari
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, P.O. box: 1475886671, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Fartoosi
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, P.O. box: 1475886671, Tehran, Iran
| | - Ahmad Fartoosi
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, P.O. box: 1475886671, Tehran, Iran
| | - Marjan Shariatpanahi
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, P.O. box: 1475886671, Tehran, Iran
| | - Mehdi Aghsami
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, P.O. box: 1475886671, Tehran, Iran
| | - Hamed Montazeri
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Afshin Kheradmand
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, P.O. box: 1475886671, Tehran, Iran.
| |
Collapse
|
35
|
Alrouji M, Al-Kuraishy HM, Al-Mahammadawy AKAA, Al-Gareeb AI, Saad HM, Batiha GES. The potential role of cholesterol in Parkinson's disease neuropathology: perpetrator or victim. Neurol Sci 2023; 44:3781-3794. [PMID: 37428278 DOI: 10.1007/s10072-023-06926-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by deposition of α-synuclein and aggregation of Lewy bodies. Cholesterol is involved with PD neuropathology in bidirectional ways that could be protective or harmful. Thus, the objective of the present review was to verify the potential role of cholesterol in PD neuropathology. Deregulation of ion channels and receptors induced by cholesterol alteration suggests a possible mechanism for the neuroprotective effects of cholesterol against PD development. However, high serum cholesterol level increases PD risk indirectly by 27-hydroxycholesterol which induces oxidative stress, inflammation, and apoptosis. Besides, hypercholesterolemia triggers the accumulation of cholesterol in macrophages and immune cells leading to the release of pro-inflammatory cytokines with progression of neuroinflammation subsequently. Additionally, cholesterol increases aggregation of α-synuclein and induces degeneration of dopaminergic neurons (DN) in the substantia nigra (SN). Hypercholesterolemia may lead to cellular Ca2+ overload causing synaptic and the development of neurodegeneration. In conclusion, cholesterol has bidirectional effects on PD neuropathology and might be protective or harmful.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, Al-Mustansiriyah University, M.B.Ch.B, FRCP; Box, Baghdad, 14132, Iraq
| | | | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, Al-Mustansiriyah University, M.B.Ch.B, FRCP; Box, Baghdad, 14132, Iraq
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matrouh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Al Beheira, 22511, Egypt.
| |
Collapse
|
36
|
Al-kuraishy HM, Alexiou A, Papadakis M, Elhussieny O, Saad HM, Batiha GES. New insights on the potential effect of vinpocetine in Parkinson's disease: one of the neglected warden and baffling topics. Metab Brain Dis 2023; 38:1831-1840. [PMID: 37335452 PMCID: PMC10348926 DOI: 10.1007/s11011-023-01254-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
Vinpocetine (VPN) is an ethyl apovincaminate that has anti-inflammatory and antioxidant effects by inhibiting the expression of nuclear factor kappa B (NF-κB) and phosphodiesterase enzyme 1 (PDE-1). VPN is used in the management of stroke, dementia, and other neurodegenerative brain diseases. VPN may be effective in treating Parkinson's disease (PD). Therefore, this review aimed to clarify the mechanistic role of VPN in the management of PD. VPN has protective and restorative effects against neuronal injury by reducing neuroinflammation, and improvement of synaptic plasticity and cerebral blood flow. VPN protects dopaminergic neurons by reducing oxidative stress, lipid peroxidation, glutamate neurotoxicity, and regulation of Ca+ 2 overloads. VPN can alleviate PD neuropathology through its anti-inflammatory, antioxidant, antiapoptotic and neurogenic effects. VPN through inhibition of PDE1 improves cyclic adenosine monophosphate (cAMP)/cyclic guanosine monophosphate (cGMP) signaling in the dopaminergic neurons of the substantia nigra (SN). VPN improves PD neuropathology through PDE1 inhibition with a subsequent increase of the cAMP/cGMP signaling pathway. Therefore, increasing cAMP leads to antioxidant effects, while augmentation of cGMP by VPN leads to anti-inflammatory effects which reduced neurotoxicity and development of motor severity in PD. In conclusion, this review indicated that VPN could be effective in the management of PD.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al- Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770 Australia
- AFNP Med, 1030 Wien, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal, Germany
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, 51744 Marsa Matruh, Egypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Egypt
| |
Collapse
|
37
|
Nelson ML, Pfeifer JA, Hickey JP, Collins AE, Kalisch BE. Exploring Rosiglitazone's Potential to Treat Alzheimer's Disease through the Modulation of Brain-Derived Neurotrophic Factor. BIOLOGY 2023; 12:1042. [PMID: 37508471 PMCID: PMC10376118 DOI: 10.3390/biology12071042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/24/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that debilitates over 55 million individuals worldwide. Currently, treatments manage and alleviate its symptoms; however, there is still a need to find a therapy that prevents or halts disease progression. Since AD has been labeled as "type 3 diabetes" due to its similarity in pathological hallmarks, molecular pathways, and comorbidity with type 2 diabetes mellitus (T2DM), there is growing interest in using anti-diabetic drugs for its treatment. Rosiglitazone (RSG) is a peroxisome proliferator-activated receptor-gamma agonist that reduces hyperglycemia and hyperinsulinemia and improves insulin signaling. In cellular and rodent models of T2DM-associated cognitive decline and AD, RSG has been reported to improve cognitive impairment and reverse AD-like pathology; however, results from human clinical trials remain consistently unsuccessful. RSG has also been reported to modulate the expression of brain-derived neurotrophic factor (BDNF), a protein that regulates neuroplasticity and energy homeostasis and is implicated in both AD and T2DM. The present review investigates RSG's limitations and potential therapeutic benefits in pre-clinical models of AD through its modulation of BDNF expression.
Collapse
Affiliation(s)
- Mackayla L Nelson
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Julia A Pfeifer
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jordan P Hickey
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Andrila E Collins
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Bettina E Kalisch
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
38
|
Evancho A, Tyler WJ, McGregor K. A review of combined neuromodulation and physical therapy interventions for enhanced neurorehabilitation. Front Hum Neurosci 2023; 17:1151218. [PMID: 37545593 PMCID: PMC10400781 DOI: 10.3389/fnhum.2023.1151218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/30/2023] [Indexed: 08/08/2023] Open
Abstract
Rehabilitation approaches for individuals with neurologic conditions have increasingly shifted toward promoting neuroplasticity for enhanced recovery and restoration of function. This review focuses on exercise strategies and non-invasive neuromodulation techniques that target neuroplasticity, including transcranial magnetic stimulation (TMS), vagus nerve stimulation (VNS), and peripheral nerve stimulation (PNS). We have chosen to focus on non-invasive neuromodulation techniques due to their greater potential for integration into routine clinical practice. We explore and discuss the application of these interventional strategies in four neurological conditions that are frequently encountered in rehabilitation settings: Parkinson's Disease (PD), Traumatic Brain Injury (TBI), stroke, and Spinal Cord Injury (SCI). Additionally, we discuss the potential benefits of combining non-invasive neuromodulation with rehabilitation, which has shown promise in accelerating recovery. Our review identifies studies that demonstrate enhanced recovery through combined exercise and non-invasive neuromodulation in the selected patient populations. We primarily focus on the motor aspects of rehabilitation, but also briefly address non-motor impacts of these conditions. Additionally, we identify the gaps in current literature and barriers to implementation of combined approaches into clinical practice. We highlight areas needing further research and suggest avenues for future investigation, aiming to enhance the personalization of the unique neuroplastic responses associated with each condition. This review serves as a resource for rehabilitation professionals and researchers seeking a comprehensive understanding of neuroplastic exercise interventions and non-invasive neuromodulation techniques tailored for specific diseases and diagnoses.
Collapse
Affiliation(s)
- Alexandra Evancho
- Department of Physical Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, United States
| | - William J. Tyler
- Department of Biomedical Engineering, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Physical Medicine and Rehabilitation, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Keith McGregor
- Department of Clinical and Diagnostic Studies, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
39
|
da Cunha Germano BC, de Morais LCC, Idalina Neta F, Fernandes ACL, Pinheiro FI, do Rego ACM, Araújo Filho I, de Azevedo EP, de Paiva Cavalcanti JRL, Guzen FP, Cobucci RN. Vitamin E and Its Molecular Effects in Experimental Models of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:11191. [PMID: 37446369 DOI: 10.3390/ijms241311191] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
With the advancement of in vivo studies and clinical trials, the pathogenesis of neurodegenerative diseases has been better understood. However, gaps still need to be better elucidated, which justifies the publication of reviews that explore the mechanisms related to the development of these diseases. Studies show that vitamin E supplementation can protect neurons from the damage caused by oxidative stress, with a positive impact on the prevention and progression of neurodegenerative diseases. Thus, this review aims to summarize the scientific evidence of the effects of vitamin E supplementation on neuroprotection and on neurodegeneration markers in experimental models. A search for studies published between 2000 and 2023 was carried out in the PubMed, Web of Science, Virtual Health Library (BVS), and Embase databases, in which the effects of vitamin E in experimental models of neurodegeneration were investigated. A total of 5669 potentially eligible studies were identified. After excluding the duplicates, 5373 remained, of which 5253 were excluded after checking the titles, 90 articles after reading the abstracts, and 11 after fully reviewing the manuscripts, leaving 19 publications to be included in this review. Experiments with in vivo models of neurodegenerative diseases demonstrated that vitamin E supplementation significantly improved memory, cognition, learning, motor function, and brain markers associated with neuroregeneration and neuroprotection. Vitamin E supplementation reduced beta-amyloid (Aβ) deposition and toxicity in experimental models of Alzheimer's disease. In addition, it decreased tau-protein hyperphosphorylation and increased superoxide dismutase and brain-derived neurotrophic factor (BDNF) levels in rodents, which seems to indicate the potential use of vitamin E in preventing and delaying the progress of degenerative lesions in the central nervous system.
Collapse
Affiliation(s)
- Bianca Caroline da Cunha Germano
- Postgraduate Program in Science Applied to Women's Health, Federal University of Rio Grande do Norte (UFRN), Natal 59072-970, Brazil
| | - Lara Cristina Carlos de Morais
- Postgratuate Program in Health and Society, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
| | - Francisca Idalina Neta
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
- Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
| | - Amélia Carolina Lopes Fernandes
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
- Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
| | - Francisco Irochima Pinheiro
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal 59056-000, Brazil
- Medical School, Health School, Potiguar University (UnP), Natal 59056-000, Brazil
| | | | - Irami Araújo Filho
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal 59056-000, Brazil
- Medical School, Health School, Potiguar University (UnP), Natal 59056-000, Brazil
| | - Eduardo Pereira de Azevedo
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal 59056-000, Brazil
| | - José Rodolfo Lopes de Paiva Cavalcanti
- Postgratuate Program in Health and Society, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
- Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
| | - Fausto Pierdona Guzen
- Postgratuate Program in Health and Society, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
- Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró 59607-360, Brazil
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal 59056-000, Brazil
| | - Ricardo Ney Cobucci
- Postgraduate Program in Science Applied to Women's Health, Federal University of Rio Grande do Norte (UFRN), Natal 59072-970, Brazil
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal 59056-000, Brazil
- Medical School, Health School, Potiguar University (UnP), Natal 59056-000, Brazil
- Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal 59078-900, Brazil
| |
Collapse
|
40
|
Félix-Soriano E, Stanford KI. Exerkines and redox homeostasis. Redox Biol 2023; 63:102748. [PMID: 37247469 PMCID: PMC10236471 DOI: 10.1016/j.redox.2023.102748] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/31/2023] Open
Abstract
Exercise physiology has gained increasing interest due to its wide effects to promote health. Recent years have seen a growth in this research field also due to the finding of several circulating factors that mediate the effects of exercise. These factors, termed exerkines, are metabolites, growth factors, and cytokines secreted by main metabolic organs during exercise to regulate exercise systemic and tissue-specific effects. The metabolic effects of exerkines have been broadly explored and entail a promising target to modulate beneficial effects of exercise in health and disease. However, exerkines also have broad effects to modulate redox signaling and homeostasis in several cellular processes to improve stress response. Since redox biology is central to exercise physiology, this review summarizes current evidence for the cross-talk between redox biology and exerkines actions. The role of exerkines in redox biology entails a response to oxidative stress-induced pathological cues to improve health outcomes and to modulate exercise adaptations that integrate redox signaling.
Collapse
Affiliation(s)
- Elisa Félix-Soriano
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
41
|
Hwang E, Portillo B, Grose K, Fujikawa T, Williams KW. Exercise-induced hypothalamic neuroplasticity: Implications for energy and glucose metabolism. Mol Metab 2023; 73:101745. [PMID: 37268247 PMCID: PMC10326746 DOI: 10.1016/j.molmet.2023.101745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/15/2023] [Accepted: 05/29/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Neuroplasticity refers to the brain's ability to undergo functional and structural changes in response to diverse challenges. Converging evidence supports the notion that exercise serves as a metabolic challenge, triggering the release of multiple factors both in the periphery and within the brain. These factors actively contribute to plasticity in the brain, and in turn, regulate energy and glucose metabolism. SCOPE OF REVIEW The primary focus of this review is to explore the impact of exercise-induced plasticity in the brain on metabolic homeostasis, with an emphasis on the role of the hypothalamus in this process. Additionally, the review provides an overview of various factors induced by exercise that contribute to energy balance and glucose metabolism. Notably, these factors exert their effects, at least in part, through actions within the hypothalamus and more broadly in the central nervous system. MAJOR CONCLUSIONS Exercise elicits both transient and sustained changes in metabolism, accompanied by changes in neural activity within specific brain regions. Importantly, the contribution of exercise-induced plasticity and the underlying mechanisms by which neuroplasticity influences the effects of exercise are not well understood. Recent work has begun to overcome this gap in knowledge by examining the complex interactions of exercise-induced factors which alter neural circuit properties to influence metabolism.
Collapse
Affiliation(s)
- Eunsang Hwang
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Bryan Portillo
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kyle Grose
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Teppei Fujikawa
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
42
|
Perl YS, Zamora-Lopez G, Montbrió E, Monge-Asensio M, Vohryzek J, Fittipaldi S, Campo CG, Moguilner S, Ibañez A, Tagliazucchi E, Yeo BTT, Kringelbach ML, Deco G. The impact of regional heterogeneity in whole-brain dynamics in the presence of oscillations. Netw Neurosci 2023; 7:632-660. [PMID: 37397876 PMCID: PMC10312285 DOI: 10.1162/netn_a_00299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/02/2022] [Indexed: 12/25/2023] Open
Abstract
Large variability exists across brain regions in health and disease, considering their cellular and molecular composition, connectivity, and function. Large-scale whole-brain models comprising coupled brain regions provide insights into the underlying dynamics that shape complex patterns of spontaneous brain activity. In particular, biophysically grounded mean-field whole-brain models in the asynchronous regime were used to demonstrate the dynamical consequences of including regional variability. Nevertheless, the role of heterogeneities when brain dynamics are supported by synchronous oscillating state, which is a ubiquitous phenomenon in brain, remains poorly understood. Here, we implemented two models capable of presenting oscillatory behavior with different levels of abstraction: a phenomenological Stuart-Landau model and an exact mean-field model. The fit of these models informed by structural- to functional-weighted MRI signal (T1w/T2w) allowed us to explore the implication of the inclusion of heterogeneities for modeling resting-state fMRI recordings from healthy participants. We found that disease-specific regional functional heterogeneity imposed dynamical consequences within the oscillatory regime in fMRI recordings from neurodegeneration with specific impacts on brain atrophy/structure (Alzheimer's patients). Overall, we found that models with oscillations perform better when structural and functional regional heterogeneities are considered, showing that phenomenological and biophysical models behave similarly at the brink of the Hopf bifurcation.
Collapse
Affiliation(s)
- Yonatan Sanz Perl
- Department of Physics, University of Buenos Aires, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), CABA, Buenos Aires, Argentina
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- Center for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Gorka Zamora-Lopez
- Center for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ernest Montbrió
- Neuronal Dynamics Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Martí Monge-Asensio
- Center for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jakub Vohryzek
- Center for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu Fabra, Barcelona, Spain
- Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, United Kingdom
| | - Sol Fittipaldi
- National Scientific and Technical Research Council (CONICET), CABA, Buenos Aires, Argentina
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- Global Brain Health Institute, University of California, San Francisco, CA, USA; and Trinity College Dublin, Dublin, Ireland
| | - Cecilia González Campo
- National Scientific and Technical Research Council (CONICET), CABA, Buenos Aires, Argentina
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
| | - Sebastián Moguilner
- Global Brain Health Institute, University of California, San Francisco, CA, USA; and Trinity College Dublin, Dublin, Ireland
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Agustín Ibañez
- National Scientific and Technical Research Council (CONICET), CABA, Buenos Aires, Argentina
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- Global Brain Health Institute, University of California, San Francisco, CA, USA; and Trinity College Dublin, Dublin, Ireland
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - Enzo Tagliazucchi
- Department of Physics, University of Buenos Aires, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), CABA, Buenos Aires, Argentina
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
| | - B. T. Thomas Yeo
- Centre for Sleep and Cognition, Centre for Translational MR Research, Department of Electrical and Computer Engineering, N.1 Institute for Health and Institute for Digital Medicine, National University of Singapore, Singapore
| | - Morten L. Kringelbach
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Center for Music in the Brain, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, United Kingdom
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu Fabra, Barcelona, Spain
- Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
- Institució Catalana de la Recerca i Estudis Avancats (ICREA), Barcelona, Spain
- Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- School of Psychological Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
43
|
Sanz Perl Y, Fittipaldi S, Gonzalez Campo C, Moguilner S, Cruzat J, Fraile-Vazquez ME, Herzog R, Kringelbach ML, Deco G, Prado P, Ibanez A, Tagliazucchi E. Model-based whole-brain perturbational landscape of neurodegenerative diseases. eLife 2023; 12:e83970. [PMID: 36995213 PMCID: PMC10063230 DOI: 10.7554/elife.83970] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
The treatment of neurodegenerative diseases is hindered by lack of interventions capable of steering multimodal whole-brain dynamics towards patterns indicative of preserved brain health. To address this problem, we combined deep learning with a model capable of reproducing whole-brain functional connectivity in patients diagnosed with Alzheimer's disease (AD) and behavioral variant frontotemporal dementia (bvFTD). These models included disease-specific atrophy maps as priors to modulate local parameters, revealing increased stability of hippocampal and insular dynamics as signatures of brain atrophy in AD and bvFTD, respectively. Using variational autoencoders, we visualized different pathologies and their severity as the evolution of trajectories in a low-dimensional latent space. Finally, we perturbed the model to reveal key AD- and bvFTD-specific regions to induce transitions from pathological to healthy brain states. Overall, we obtained novel insights on disease progression and control by means of external stimulation, while identifying dynamical mechanisms that underlie functional alterations in neurodegeneration.
Collapse
Affiliation(s)
- Yonatan Sanz Perl
- Department of Physics, University of Buenos AiresBuenos AiresArgentina
- National Scientific and Technical Research Council (CONICET), CABABuenos AiresArgentina
- Cognitive Neuroscience Center (CNC), Universidad de San AndrésBuenos AiresArgentina
- Center for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu FabraBarcelonaSpain
| | - Sol Fittipaldi
- National Scientific and Technical Research Council (CONICET), CABABuenos AiresArgentina
- Cognitive Neuroscience Center (CNC), Universidad de San AndrésBuenos AiresArgentina
| | - Cecilia Gonzalez Campo
- National Scientific and Technical Research Council (CONICET), CABABuenos AiresArgentina
- Cognitive Neuroscience Center (CNC), Universidad de San AndrésBuenos AiresArgentina
| | - Sebastián Moguilner
- Global Brain Health Institute, University of California, San FranciscoSan FranciscoUnited States
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo IbáñezSantiagoChile
| | - Josephine Cruzat
- Center for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu FabraBarcelonaSpain
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo IbáñezSantiagoChile
| | | | - Rubén Herzog
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo IbáñezSantiagoChile
| | - Morten L Kringelbach
- Department of Psychiatry, University of OxfordOxfordUnited Kingdom
- Center for Music in the Brain, Department of Clinical Medicine, Aarhus UniversityÅrhusDenmark
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of MinhoBragaPortugal
- Centre for Eudaimonia and Human Flourishing, University of OxfordOxfordUnited Kingdom
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu FabraBarcelonaSpain
- Department of Information and Communication Technologies, Universitat Pompeu FabraBarcelonaSpain
- Institució Catalana de la Recerca i Estudis Avancats (ICREA)BarcelonaSpain
- Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
- School of Psychological Sciences, Monash UniversityClaytonAustralia
| | - Pavel Prado
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo IbáñezSantiagoChile
- Escuela de Fonoaudiología, Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San SebastiánSantiagoChile
| | - Agustin Ibanez
- National Scientific and Technical Research Council (CONICET), CABABuenos AiresArgentina
- Cognitive Neuroscience Center (CNC), Universidad de San AndrésBuenos AiresArgentina
- Global Brain Health Institute, University of California, San FranciscoSan FranciscoUnited States
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo IbáñezSantiagoChile
- Trinity College Institute of Neuroscience (TCIN), Trinity College DublinDublinIreland
| | - Enzo Tagliazucchi
- Department of Physics, University of Buenos AiresBuenos AiresArgentina
- National Scientific and Technical Research Council (CONICET), CABABuenos AiresArgentina
- Cognitive Neuroscience Center (CNC), Universidad de San AndrésBuenos AiresArgentina
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo IbáñezSantiagoChile
| |
Collapse
|
44
|
Saral S, Topçu A, Alkanat M, Mercantepe T, Şahin Z, Akyıldız K, Karataş KS, Yıldız L, Tümkaya L, Yazıcı ZA. Agomelatine attenuates cisplatin-induced cognitive impairment via modulation of BDNF/TrkB signaling in rat hippocampus. J Chem Neuroanat 2023; 130:102269. [PMID: 37001681 DOI: 10.1016/j.jchemneu.2023.102269] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
Cisplatin is a drug used effectively in the treatment of malignant tumors. However, cisplatin has many side effects, including cognitive impairment. Agomelatine, a synthetic melatonin analogue, is an important antidepressant. Increasing evidence has shown that agomelatine may be a potential neuroprotective agent. The aim of this study was to investigate the effect of agomelatine on learning and memory functions in cisplatin-induced cognitive impairment in a rat model. Male rats were administered agomelatine and cisplatin for 4 weeks. Neurobehavioral tests were performed at the end of the 4th week. After behavioral tests, rats were euthanized and BDNF, TNF, IL-1β, MDA and GSH levels were measured in hippocampal homegenates by ELISA. In addition, nNOS and TrkB receptor activity were measured immunohistochemically. The results showed that agomelatine significantly improved cognitive functions in spatial memory tests in rats with cisplatin-induced cognitive impairment. In addition, agomelatine treatment positively affected the discrimination index (DI). On the other hand, agomelatine treatment elevated cisplatin-suppressed hippocampal BDNF levels. Agomelatine treatment reduced cisplatin-induced neuroinflammation by suppressing TNF and IL-1β levels. Similarly, agomelatine reduced oxidative stress in the hippocampus. Histological findings showed that agomelatine treatment reduced pyramidal neuron damage in hippocampal DG, CA1 and CA3. Cisplatin increased nNOS and TrkB positivity in DG, CA1 and CA3 neurons compared to control. In contrast, agomelatine treatment decreased both nNOS and TrkB positive scores. These findings indicate that agomelatine reduces cisplatin-related cognitive impairment by exerting anti-inflammatory action and possibly by the modulation of the BDNF/TrkB/nNOS pathways in the hippocampus.
Collapse
Affiliation(s)
- Sinan Saral
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Physiology, Rize, Turkey.
| | - Atilla Topçu
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Pharmacology, Rize, Turkey
| | - Mehmet Alkanat
- Giresun University, Faculty of Medicine, Department of Physiology, Giresun, Turkey
| | - Tolga Mercantepe
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Histology and Embryology, Rize, Turkey
| | - Zafer Şahin
- Karadeniz Technical University, Faculty of Medicine, Department of Physiology, Trabzon, Turkey
| | - Kerimali Akyıldız
- Recep Tayyip Erdogan University, School of Healh Care Services Vocational, Department of Medical Services and Techniques, Rize, Turkey
| | - Kader Semra Karataş
- Kutahya Health Sciences of University, Faculty of Medicine, Department of Mental Health and Diseases, Kütahya, Turkey
| | - Lamiye Yıldız
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Physiology, Rize, Turkey
| | - Levent Tümkaya
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Histology and Embryology, Rize, Turkey
| | - Zihni Açar Yazıcı
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Microbiology, Rize, Turkey
| |
Collapse
|
45
|
Sharma V, Singh TG, Kaur A, Mannan A, Dhiman S. Brain-Derived Neurotrophic Factor: A Novel Dynamically Regulated Therapeutic Modulator in Neurological Disorders. Neurochem Res 2023; 48:317-339. [PMID: 36308619 DOI: 10.1007/s11064-022-03755-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 02/04/2023]
Abstract
The growth factor brain-derived neurotrophic factor (BDNF), and its receptor tropomyosin-related kinase receptor type B (TrkB) play an active role in numerous areas of the adult brain, where they regulate the neuronal activity, function, and survival. Upregulation and downregulation of BDNF expression are critical for the physiology of neuronal circuits and functioning in the brain. Loss of BDNF function has been reported in the brains of patients with neurodegenerative or psychiatric disorders. This article reviews the BDNF gene structure, transport, secretion, expression and functions in the brain. This article also implicates BDNF in several brain-related disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, major depressive disorder, schizophrenia, epilepsy and bipolar disorder.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India.
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| |
Collapse
|
46
|
Ramos JS, Chakraborty R, Dalleck LC, Sarunic K, Khadka J, Haslam T, Nassaris O. A retrospective evaluation of the Brain and Body Fitness Studio service on functional capacity and quality of life in people with neurological disorders. Front Neurol 2023; 13:1006221. [PMID: 36742042 PMCID: PMC9895383 DOI: 10.3389/fneur.2022.1006221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/21/2022] [Indexed: 01/22/2023] Open
Abstract
Background People with neurological disorders (ND) are less physically active than the general population due to physical, sensory, and/or cognitive impairments. These individuals often feel intimidated to join mainstream health and wellness centers due to lack of specialized support for people with ND. The Brain and Body Fitness Studio (BBFS) is one of the first Accredited Exercise Physiologist-led interprofessional services in Adelaide South Australia to provide individualized evidence-based multimodal exercise prescription and social support for this population. This comprehensive retrospective study evaluated the impact of BBFS on functional capacity (FC) determined as the 6-min walk distance (6 MWD) achieved during a 6-min walk test (6 MWT), of its members with ND. Methods Sixty-two BBFS members (age, 66 ± 10 years; 60% male) with ND (85% Parkinson's Disease; average time since diagnosis, 4 years [IQR, 2 to 12 years]) and complete pre- and post-6-month clinical assessment of the primary outcome of the study, the 6 MWD, were included in this retrospective analysis. A series of sub-analyses were also performed to investigate the effects of adherence to the recommended prescription of at least twice a week in the program (≥80 vs. < 80% adherence), and disease stage (time since diagnosis; ≥6 vs. < 6 years) on FC. Results Although there was no statistically significant change in 6 MWD from pre- to post-6-month BBFS program (+15 ± 90 m, p = 0.19), a clinically meaningful improvement of >14 m was evident. Improvement in 6 MWD was significantly greater in members who attended at least 80% of the recommended visits (≥80% visits, +37 ± 58 m; ≤ 80% visits,-1 ± 105 m, p = 0.046). We also found a 6 MWD improvement from pre- to post-6 months in those in the early years of their ND (< 6 years since diagnosis, +39 ± 76 m), but not in those in the later years of their ND (≥6 years since diagnosis, -36 ± 123 m, between group difference, p = 0.029). Conclusion A clinically meaningful 6 MWD improvement may be elicited by services provided by BBFS in people with ND. Overall, the benefits appear to be more evident in members who attended the BBFS for at least 80% of the recommended visits and those who were in the early stage of their ND diagnosis.
Collapse
Affiliation(s)
- Joyce S. Ramos
- Clinical Exercise Physiology, College of Nursing and Health Sciences, SHAPE Research Centre, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Ranjay Chakraborty
- Clinical Exercise Physiology, College of Nursing and Health Sciences, SHAPE Research Centre, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Lance C. Dalleck
- Clinical Exercise Physiology, College of Nursing and Health Sciences, SHAPE Research Centre, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
- Recreation, Exercise and Sport, Western Colorado University, Gunnison, CO, United States
| | - Kristina Sarunic
- Clinical Exercise Physiology, College of Nursing and Health Sciences, SHAPE Research Centre, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Jyoti Khadka
- Clinical Exercise Physiology, College of Nursing and Health Sciences, SHAPE Research Centre, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
- Health and Social Care Economics Group, College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Tayla Haslam
- Brain and Body Fitness Studio, The Hospital Research Foundation Parkinson's, Adelaide, SA, Australia
| | - Olivia Nassaris
- Clinical Exercise Physiology, College of Nursing and Health Sciences, SHAPE Research Centre, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
- Brain and Body Fitness Studio, The Hospital Research Foundation Parkinson's, Adelaide, SA, Australia
| |
Collapse
|
47
|
Kouter K, Nikolac Perkovic M, Nedic Erjavec G, Milos T, Tudor L, Uzun S, Mimica N, Pivac N, Videtic Paska A. Difference in Methylation and Expression of Brain-Derived Neurotrophic Factor in Alzheimer's Disease and Mild Cognitive Impairment. Biomedicines 2023; 11:235. [PMID: 36830773 PMCID: PMC9953261 DOI: 10.3390/biomedicines11020235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Due to the increasing number of progressive dementias in the population, numerous studies are being conducted that seek to determine risk factors, biomarkers and pathological mechanisms that could help to differentiate between normal symptoms of aging, mild cognitive impairment (MCI) and dementia. The aim of this study was to investigate the possible association of levels of BDNF and COMT gene expression and methylation in peripheral blood cells with the development of Alzheimer's disease (AD). Our results revealed higher expression levels of BDNF (p < 0.001) in MCI subjects compared to individuals diagnosed with AD. However, no difference in COMT gene expression (p = 0.366) was detected. DNA methylation of the CpG islands and other sequences with potential effects on gene expression regulation revealed just one region (BDNF_9) in the BDNF gene (p = 0.078) with marginally lower levels of methylation in the AD compared to MCI subjects. Here, we show that the level of BDNF expression in the periphery is decreased in subjects with AD compared to individuals with MCI. The combined results from the gene expression analysis and DNA methylation analysis point to the potential of BDNF as a marker that could help distinguish between MCI and AD patients.
Collapse
Affiliation(s)
- Katarina Kouter
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Matea Nikolac Perkovic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Gordana Nedic Erjavec
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Tina Milos
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Lucija Tudor
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Suzana Uzun
- Department for Biological Psychiatry and Psychogeriatrics, University Psychiatric Hospital Vrapce, 10090 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ninoslav Mimica
- Department for Biological Psychiatry and Psychogeriatrics, University Psychiatric Hospital Vrapce, 10090 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Nela Pivac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Alja Videtic Paska
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
48
|
Takla M, Saadeh K, Tse G, Huang CLH, Jeevaratnam K. Ageing and the Autonomic Nervous System. Subcell Biochem 2023; 103:201-252. [PMID: 37120470 DOI: 10.1007/978-3-031-26576-1_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The vertebrate nervous system is divided into central (CNS) and peripheral (PNS) components. In turn, the PNS is divided into the autonomic (ANS) and enteric (ENS) nervous systems. Ageing implicates time-related changes to anatomy and physiology in reducing organismal fitness. In the case of the CNS, there exists substantial experimental evidence of the effects of age on individual neuronal and glial function. Although many such changes have yet to be experimentally observed in the PNS, there is considerable evidence of the role of ageing in the decline of ANS function over time. As such, this chapter will argue that the ANS constitutes a paradigm for the physiological consequences of ageing, as well as for their clinical implications.
Collapse
Affiliation(s)
| | | | - Gary Tse
- Kent and Medway Medical School, Canterbury, UK
- University of Surrey, Guildford, UK
| | | | | |
Collapse
|
49
|
Fan J, Du C, Li X. Emotional and Affective Disorders in Cognitive Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:63-71. [PMID: 37418206 DOI: 10.1007/978-981-99-1627-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
The increased aging population who have aging-related diseases poses a serious challenge to health services, including mental health services. Due to the changes of body, brain, living environment, and lifestyle, the elderly will have different psychological changes from other age stages, some of which would develop into mental disorders, and affect the cognition of the elderly in turn. This elderly mental health condition has drawn wide attention from scientists. This chapter introduces the two most common emotional and affective disorders, late-life depression and anxiety, and focuses on their epidemiology and impact on the elderly. Furthermore, this chapter also reviews the effects of these two disorders on cognitive function and cognitive impairment in the elderly, and tries to explain the underlying mechanism of this effect from the perspective of related disease, cerebral circuit, and molecular biology.
Collapse
Affiliation(s)
- Jialing Fan
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Chao Du
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Xin Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China.
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China.
| |
Collapse
|
50
|
Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Bryukhovetskiy I, Manzhulo I, Wiklund L, Sharma HS. Nanowired Delivery of Cerebrolysin Together with Antibodies to Amyloid Beta Peptide, Phosphorylated Tau, and Tumor Necrosis Factor Alpha Induces Superior Neuroprotection in Alzheimer's Disease Brain Pathology Exacerbated by Sleep Deprivation. ADVANCES IN NEUROBIOLOGY 2023; 32:3-53. [PMID: 37480458 DOI: 10.1007/978-3-031-32997-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Sleep deprivation induces amyloid beta peptide and phosphorylated tau deposits in the brain and cerebrospinal fluid together with altered serotonin metabolism. Thus, it is likely that sleep deprivation is one of the predisposing factors in precipitating Alzheimer's disease (AD) brain pathology. Our previous studies indicate significant brain pathology following sleep deprivation or AD. Keeping these views in consideration in this review, nanodelivery of monoclonal antibodies to amyloid beta peptide (AβP), phosphorylated tau (p-tau), and tumor necrosis factor alpha (TNF-α) in sleep deprivation-induced AD is discussed based on our own investigations. Our results suggest that nanowired delivery of monoclonal antibodies to AβP with p-tau and TNF-α induces superior neuroprotection in AD caused by sleep deprivation, not reported earlier.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Department Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|